1
|
Thirumoorthy C, Sharma KK, Deepa M, Yogaprabhu S, Sneha J, Rekha RP, Venkatesan U, Hemavathy S, Nikhil J, Srikumar BN, Binukumar B, Radha V, Sharma S, Grallert H, Ball G, Ram U, Anjana RM, Balasubramanyam M, Tandon N, Mohan V, Saravanan P, Gokulakrishnan K. Epigenome-wide association study identifies a specific panel of DNA methylation signatures for antenatal and postpartum depressive symptoms. J Affect Disord 2025; 383:195-206. [PMID: 40286927 DOI: 10.1016/j.jad.2025.04.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/13/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Depression during pregnancy and postpartum poses significant risks to both maternal and child well-being. The underlying biological mechanisms are unclear, but epigenetic variation could be exploited as a plausible candidate for early detection. We investigated whether DNA methylation signatures are associated with antenatal depressive symptoms (ADS) and whether early alterations in methylation patterns could be used to predict postpartum depressive symptoms (PDS). 201 pregnant women in early pregnancy, without a prior history of depressive disorders, from the STratification of Risk of Diabetes in Early Pregnancy study (STRiDE) were recruited. Using the Patient Health Questionnaire-9 (PHQ-9), 92 women were identified with ADS, while 109 served as controls. Edinburgh Postnatal Depression Scale (EPDS) was used to assess PDS during 6-12 weeks after delivery. The dataset was split into 80 % for training and testing and 20 % for validation, to discern potential CpGs for ADS using a support vector machine classifier. Analysis revealed 591 CpGs significantly associated with ADS, from which a panel of 7 CpGs was identified to discriminate between ADS and controls with high sensitivity and specificity (AUC: 0.85 in test, 0.73 in validation). Pathway analysis highlighted involvement in inositol phosphate metabolism, notch, and calcium signaling. The same 7 CpGs predicted PDS with an AUC of 0.76 (95 % CI: 0.66-0.87). Integration of CpG data with patient-reported information significantly enhanced PDS prediction. Our study identified DNA methylation signatures that could potentially differentiate ADS from controls and predict PDS. This suggests potential for developing a CpG panel for diagnostic and preventive strategies for perinatal depression.
Collapse
Affiliation(s)
- Chinnasamy Thirumoorthy
- Department of Neurochemistry, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bengaluru, India
| | - Kuldeep Kumar Sharma
- Biostatistics, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bengaluru, India
| | - Mohan Deepa
- Department of Diabetology, Madras Diabetes Research Foundation (MDRF), Chennai, India
| | - Saravanan Yogaprabhu
- Department of Molecular Genetics, Madras Diabetes Research Foundation (MDRF), Affiliated to University of Madras, Chennai, India
| | - Janaki Sneha
- Department of Molecular Genetics, Madras Diabetes Research Foundation (MDRF), Affiliated to University of Madras, Chennai, India
| | - Ravikumar Pavithra Rekha
- Department of Neurochemistry, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bengaluru, India
| | | | - Saite Hemavathy
- Department of Diabetology, Madras Diabetes Research Foundation (MDRF), Chennai, India
| | - Joyappa Nikhil
- Department of Neurochemistry, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bengaluru, India
| | - Bettadapura N Srikumar
- Neurophysiology, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bengaluru, India
| | | | - Venkatesan Radha
- Department of Molecular Genetics, Madras Diabetes Research Foundation (MDRF), Affiliated to University of Madras, Chennai, India
| | - Sapna Sharma
- Institute of Epidemiology, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) | HZM, Germany
| | - Harald Grallert
- Institute of Epidemiology, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) | HZM, Germany
| | - Graham Ball
- Medical Technology Research Centre, Anglia Ruskin University, UK
| | - Uma Ram
- Seethapathy Clinic & Hospital, Chennai, India
| | - Ranjit Mohan Anjana
- Department of Diabetology, Madras Diabetes Research Foundation (MDRF), Chennai, India
| | | | - Nikhil Tandon
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Viswanathan Mohan
- Department of Diabetology, Madras Diabetes Research Foundation (MDRF), Chennai, India
| | - Ponnusamy Saravanan
- Populations, Evidence and Technologies, Division of Health Sciences, Warwick Medical School, University of Warwick, UK; Department of Diabetes, Endocrinology and Metabolism, George Eliot Hospital, Nuneaton, UK; Centre for Global Health, University of Warwick, UK.
| | - Kuppan Gokulakrishnan
- Department of Neurochemistry, National Institute of Mental Health & Neuro Sciences (NIMHANS), Bengaluru, India.
| |
Collapse
|
2
|
Beveridge J, Montgomery A, Grossberg G. Intermittent fasting and neurocognitive disorders: What the evidence shows. J Nutr Health Aging 2025; 29:100480. [PMID: 39798403 DOI: 10.1016/j.jnha.2025.100480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
INTRODUCTION Intermittent fasting (IF) has emerged as a potential lifestyle intervention for mitigating cognitive decline and enhancing brain health in individuals with mild to major neurocognitive disorders. Unlike preventive strategies, this review evaluates IF as a therapeutic approach, focusing on its effects on neuroplasticity, inflammation, and cognitive function. METHODS A narrative review was conducted using a comprehensive PubMed search with the terms "intermittent fasting AND neurocognition" and "intermittent fasting AND neuroplasticity". Studies published in English within the last ten years involving human and animal models were included. Exclusion criteria focused on studies primarily examining mood disorders or unrelated metabolic outcomes. RESULTS Preclinical evidence demonstrates that IF enhances hippocampal neurogenesis and synaptic plasticity through pathways involving BDNF and CREB. IF also reduces neuroinflammation, as shown in animal models of Alzheimer's disease, vascular cognitive impairment, and high-fat diet-induced cognitive impairment. Human studies, though limited, suggest that regular IF may improve cognitive function and reduce markers of oxidative stress and inflammation in individuals with mild cognitive impairment. CONCLUSION Current findings highlight the therapeutic potential of IF for individuals with existing cognitive impairment. While preclinical studies provide robust evidence of neuroprotective mechanisms, human studies remain sparse and require standardization. Further clinical research is necessary to confirm long-term safety and efficacy and to refine IF protocols for broader clinical application.
Collapse
Affiliation(s)
- Jordan Beveridge
- St. Louis University Department of Psychiatry and Behavioral Neuroscience, Monteleone Hall, 1438 South Grand Boulevard, St. Louis, MO 63104, United States.
| | - Allison Montgomery
- St. Louis University Department of Psychiatry and Behavioral Neuroscience, Monteleone Hall, 1438 South Grand Boulevard, St. Louis, MO 63104, United States
| | - George Grossberg
- St. Louis University Department of Psychiatry and Behavioral Neuroscience, Monteleone Hall, 1438 South Grand Boulevard, St. Louis, MO 63104, United States
| |
Collapse
|
3
|
de la Monte SM, Tong M, Ziplow J, Mark P, Van S, Nguyen VA. Impact of Prenatal Dietary Soy on Cerebellar Neurodevelopment and Function in Experimental Fetal Alcohol Spectrum Disorder. Nutrients 2025; 17:812. [PMID: 40077682 PMCID: PMC11901751 DOI: 10.3390/nu17050812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/01/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Prenatal alcohol exposure (PAE) models can cause neurodevelopmental abnormalities like those observed in fetal alcohol spectrum disorder (FASD). Previous studies link experimental PAE effects in the brain to impaired signaling through insulin/IGF and Notch pathways that mediate neuronal survival, growth, migration, energy metabolism, and plasticity. Importantly, concurrent administration of peroxisome proliferator-activated receptor agonists or dietary soy prevented many aspects of FASD due to their insulin-sensitizing, anti-inflammatory, and antioxidant properties. Objective: To determine if dietary soy interventions during pregnancy would be sufficient to normalize central nervous system structure and function, we examined the effects of maternal gestation-limited dietary soy on cerebellar postnatal development, motor function, and critical signaling pathways. Methods: Pregnant Long Evans rats were fed isocaloric liquid diets containing 0% or 26% caloric ethanol with casein or soy isolate as the protein source. The ethanol and soy feedings were discontinued upon delivery. The offspring were subjected to rotarod motor function tests, and on postnatal day 35, they were sacrificed to harvest cerebella for histological and molecular studies. Results: Despite the postnatal cessation of alcohol exposure, chronic gestational exposure reduced brain weight, caused cerebellar hypoplasia, and impaired motor performance. Gestational dietary soy prevented the ethanol-associated reduction in brain weight and largely restored the histological integrity of the cerebellum but failed to normalize motor performance. Ethanol withdrawal abolished the impairments in insulin/IGF signaling that were previously associated with ongoing ethanol exposures, but ethanol's inhibitory effects on Notch and Wnt signaling persisted. Soy significantly increased cerebellar expression of the insulin and IGF-1 receptors and abrogated several ethanol-associated impairments in Notch and Wnt signaling. Conclusions: Although gestation-restricted dietary soy has significant positive effects on neurodevelopment, optimum prevention of FASD's long-term effects will likely require dietary soy intervention during the critical periods of postnatal development, even after alcohol exposures have ceased.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology and Laboratory Medicine, Neurosurgery, and Neurology, Rhode Island Hospital, Providence, RI 02903, USA
- Women & Infants Hospital, Brown University Health, Providence, RI 02905, USA
- Alpert Medical School of Brown University, Providence, RI 02903, USA
- Department of Medicine, Rhode Island Hospital, Brown University Health, Providence, RI 02903, USA
| | - Ming Tong
- Alpert Medical School of Brown University, Providence, RI 02903, USA
- Department of Medicine, Rhode Island Hospital, Brown University Health, Providence, RI 02903, USA
| | - Jason Ziplow
- Departments of Neuroscience and Biology, Brown University, Providence, RI 02903, USA; (J.Z.); (S.V.)
| | - Princess Mark
- Department of Medicine, Rhode Island Hospital, Brown University Health, Providence, RI 02903, USA
| | - Stephanie Van
- Departments of Neuroscience and Biology, Brown University, Providence, RI 02903, USA; (J.Z.); (S.V.)
| | - Van Ahn Nguyen
- Departments of Neuroscience and Biology, Brown University, Providence, RI 02903, USA; (J.Z.); (S.V.)
| |
Collapse
|
4
|
Gu C, Wang Z, Luo W, Ling H, Cui X, Deng T, Li K, Huang W, Xie Q, Tao B, Qi X, Peng X, Ding J, Qiu P. Impaired olfactory bulb neurogenesis mediated by Notch1 contributes to olfactory dysfunction in mice chronically exposed to methamphetamine. Cell Biol Toxicol 2025; 41:46. [PMID: 39976779 PMCID: PMC11842540 DOI: 10.1007/s10565-025-10004-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
Methamphetamine (Meth) is a potent central nervous system stimulant with high addictive potential and neurotoxic effects. Chronic use results in significant damage in various brain functions, including cognition, memory, and sensory perception. Olfactory dysfunction is a notable yet often overlooked consequence of Meth abuse, and its underlying mechanisms are not fully understood. This study investigates the mechanisms of Meth-induced olfactory impairment through a thorough examination of olfactory bulb (OB) neurogenesis. We found that chronic Meth abuse impaired olfactory function in mice by not only reducing the self-renewal of subventricular zone (SVZ) neural stem cells (NSCs) but also altering their differentiation potential, leading their differentiation into astrocytes at the expense of neurons. Mechanistically, Meth inhibits autophagosome-lysosome fusion by downregulating Syntaxin 17 (Stx17), which reduces autophagic flux. In NSCs, autophagy tightly regulates Notch1 levels, and impaired autophagic degradation of Notch1 leads to its abnormal activation. This alters NSCs fate determination, ultimately affecting OB neurogenesis. Our study reveals that Meth impairs olfactory function through autophagic dysfunction and aberrant Notch1 signaling. Understanding these mechanisms not only provides new insights into Meth-induced olfactory dysfunction but also offers potential targets for developing therapies to alleviate Meth-induced neurotoxicity and sensory damage in the future.
Collapse
Affiliation(s)
- Cihang Gu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhuo Wang
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wenyu Luo
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Haosen Ling
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xilie Cui
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Tongtong Deng
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Kuan Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Wei Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qiqian Xie
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Bowen Tao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, 550004, China
| | - Xiaojia Peng
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jiuyang Ding
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, 550004, China.
| | - Pingming Qiu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515, China.
- Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
5
|
Toupenet Marchesi L, Stockholm D, Esteves T, Leblanc M, Auger N, Branchu J, El Hachimi KH, Stevanin G. Transcriptomic analysis reinforces the implication of spatacsin in neuroinflammation and neurodevelopment. Sci Rep 2025; 15:2370. [PMID: 39827309 PMCID: PMC11743199 DOI: 10.1038/s41598-025-86337-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Hereditary spastic paraplegia (HSP) encompasses a group of rare genetic diseases primarily affecting motor neurons. Among these, spastic paraplegia type 11 (SPG11) represents a complex form of HSP caused by deleterious variants in the SPG11 gene, which encodes the spatacsin protein. Previous studies have described several potential roles for spatacsin, including its involvement in lysosome and autophagy mechanisms, neuronal and neurites development or mitochondria function. Despite these findings, the precise function of the spatacsin protein remains elusive. To elucidate its function, we conducted an extensive RNA sequencing (RNAseq) experiment and transcriptomic analysis in three distinct neural structures (cerebellum, cortex and hippocampus) and at three different ages (6 weeks, 4 months and 8 months) in both wild type and Spg11-/- mice. Our functional analysis of differentially expressed genes (DEGs) and Gene Set Enrichment Analysis (GSEA) revealed dysregulation in pathways related to inflammation, RNA metabolism and neuronal and neurite development, factors frequently implicated in neurodegenerative disorders. Notably, we also observed early deregulation in cellular pathways related to cell proliferation. Our results represent a significant step towards a better understanding of the functions of spatacsin in the cell and the underlying cellular mechanisms disrupted by its absence.
Collapse
Affiliation(s)
- Liriopé Toupenet Marchesi
- Paris Brain Institute (ICM), Sorbonne University, INSERM, CNRS, APHP, Paris, France
- PSL Research University, EPHE, Paris, France
| | - Daniel Stockholm
- PSL Research University, EPHE, Paris, France
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Typhaine Esteves
- Paris Brain Institute (ICM), Sorbonne University, INSERM, CNRS, APHP, Paris, France
- PSL Research University, EPHE, Paris, France
| | - Marion Leblanc
- Paris Brain Institute (ICM), Sorbonne University, INSERM, CNRS, APHP, Paris, France
- PSL Research University, EPHE, Paris, France
| | - Nicolas Auger
- Paris Brain Institute (ICM), Sorbonne University, INSERM, CNRS, APHP, Paris, France
- PSL Research University, EPHE, Paris, France
| | - Julien Branchu
- Paris Brain Institute (ICM), Sorbonne University, INSERM, CNRS, APHP, Paris, France
| | - Khalid Hamid El Hachimi
- Paris Brain Institute (ICM), Sorbonne University, INSERM, CNRS, APHP, Paris, France
- PSL Research University, EPHE, Paris, France
| | - Giovanni Stevanin
- PSL Research University, EPHE, Paris, France.
- Institut des Neurosciences cognitives et intégratives d'Aquitaine (INCIA), Bordeaux University, CNRS, Bordeaux, France.
| |
Collapse
|
6
|
Abidi SNF, Chan S, Seidel K, Lafkas D, Vermeulen L, Peale F, Siebel CW. The Jag2/Notch1 signaling axis promotes sebaceous gland differentiation and controls progenitor proliferation. eLife 2024; 13:RP98747. [PMID: 39585329 PMCID: PMC11588336 DOI: 10.7554/elife.98747] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024] Open
Abstract
The sebaceous gland (SG) is a vital appendage of the epidermis, and its normal homeostasis and function is crucial for effective maintenance of the skin barrier. Notch signaling is a well-known regulator of epidermal differentiation, and has also been shown to be involved in postnatal maintenance of SGs. However, the precise role of Notch signaling in regulating SG differentiation in the adult homeostatic skin remains unclear. While there is evidence to suggest that Notch1 is the primary Notch receptor involved in regulating the differentiation process, the ligand remains unknown. Using monoclonal therapeutic antibodies designed to specifically inhibit of each of the Notch ligands or receptors, we have identified the Jag2/Notch1 signaling axis as the primary regulator of sebocyte differentiation in mouse homeostatic skin. Mature sebocytes are lost upon specific inhibition of the Jag2 ligand or Notch1 receptor, resulting in the accumulation of proliferative stem/progenitor cells in the SG. Strikingly, this phenotype is reversible, as these stem/progenitor cells re-enter differentiation when the inhibition of Notch activity is lifted. Thus, Notch activity promotes correct sebocyte differentiation, and is required to restrict progenitor proliferation.
Collapse
Affiliation(s)
| | - Sara Chan
- Department of Research Pathology, GenentechSan FranciscoUnited States
| | - Kerstin Seidel
- Department of Discovery Oncology, GenentechSan FranciscoUnited States
| | - Daniel Lafkas
- Department of Discovery Oncology, GenentechSan FranciscoUnited States
| | - Louis Vermeulen
- Department of Discovery Oncology, GenentechSan FranciscoUnited States
| | - Frank Peale
- Department of Research Pathology, GenentechSan FranciscoUnited States
| | | |
Collapse
|
7
|
Levy I, Arvidson R. Cephalic ganglia transcriptomics of the American cockroach Periplaneta americana (Blattodea: Blattidae). JOURNAL OF INSECT SCIENCE (ONLINE) 2024; 24:12. [PMID: 39688382 DOI: 10.1093/jisesa/ieae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024]
Abstract
The American cockroach Periplaneta americana (L.) (Blattodea, Blattidae) has been a model organism for biochemical and physiological study for almost a century, however, its use does not benefit from the genetic tools found in key model species such as Drosophila melanogaster. To facilitate the use of the cockroach as a model system in neuroscience and to serve as a foundation for functional and translational experimentation, a transcriptome of the cephalic ganglia was assembled and annotated, and differential expression profiles between these ganglia were assessed. The transcriptome assembly yielded >400 k transcripts, with >40 k putative coding sequences. Gene ontology and protein domain searches indicate the cerebral and gnathal ganglia (GNG) have distinct genetic expression profiles. The developmental Toll signaling pathway appears to be active in the adult central nervous system (CNS), which may suggest a separate role for this pathway besides innate immune activation or embryonic development. The catabolic glycolytic and citric acid cycle enzymes are well represented in both ganglia, but key enzymes are more highly expressed in the GNG. Both ganglia express gluconeogenic and trehaloneogenic enzymes, suggesting a larger role of the CNS in regulating hemolymph sugar homeostasis than previously appreciated. The annotation and quantification of the cephalic ganglia transcriptome reveal both canonical and novel pathways in signaling and metabolism in an adult insect and lay a foundation for future functional and genetic analysis.
Collapse
Affiliation(s)
- Ilana Levy
- Undergraduate Program in Biochemistry, Department of Biochemistry, Case Western Reserve University, Cleveland, OH, USA
| | - Ryan Arvidson
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
8
|
Mohammadi A, Balduini W, Carloni S. Melatonin modulates the Notch1 signaling pathway and Sirt3 in the hippocampus of hypoxic-ischemic neonatal rats. Sci Rep 2024; 14:25069. [PMID: 39443594 PMCID: PMC11500095 DOI: 10.1038/s41598-024-76307-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
The Notch1 signaling pathway plays a crucial role in the development of the central nervous system, governing pivotal functional activities in the brain, such as neurogenesis. Sirt3 is instrumental in managing mitochondrial homeostasis and is essential to cell survival. Dysregulation of these signaling pathways is implicated in the pathogenesis of a wide range of diseases, including neurodegenerative disorders such as stroke. We have previously shown that melatonin significantly improved the perinatal brain damage caused by hypoxia-ischemia (HI) through the activation of several protective mechanisms such as restoring mitochondria status and increasing the hippocampal cell proliferation. This study assessed whether melatonin affects the Notch1 signaling pathway and Sirt3 after neonatal HI. Results show that HI significantly increased Notch1 expression both in hippocampal neurons and glial cells as well as the expression of the key proteins of the pathway NICD, HES1, and c-Myc. Melatonin significantly prevented the Notch1 signaling pathway activation induced by HI, maintaining NICD and HES1 expression to control levels. In the same neurons, melatonin also prevents the Sirt3 depletion caused by HI. In summary, this study provides new insights into the effects of melatonin on the Notch1 signaling pathway and Sirt3 in in vivo neonatal brain ischemia. We suggest that the rapid modulation of the Notch1 signaling pathway and Sirt3 induced by melatonin may support neuronal survival during ischemia.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via S. Chiara 27, 61029, Urbino (PU), Italy
| | - Water Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via S. Chiara 27, 61029, Urbino (PU), Italy
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via S. Chiara 27, 61029, Urbino (PU), Italy.
| |
Collapse
|
9
|
Yang HH, Han KM, Kim A, Kang Y, Tae WS, Han MR, Ham BJ. Neuroimaging and epigenetic analysis reveal novel epigenetic loci in major depressive disorder. Psychol Med 2024; 54:2585-2598. [PMID: 38721773 DOI: 10.1017/s0033291724000709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
BACKGROUND Epigenetic modifications, such as DNA methylation, contribute to the pathophysiology of major depressive disorder (MDD). This study aimed to identify novel MDD-associated epigenetic loci using DNA methylation profiles and explore the correlations between epigenetic loci and cortical thickness changes in patients with MDD. METHODS A total of 350 patients with MDD and 161 healthy controls (HCs) were included in the epigenome-wide association studies (EWAS). We analyzed methylation, copy number alteration (CNA), and gene network profiles in the MDD group. A total of 234 patients with MDD and 135 HCs were included in neuroimaging methylation analysis. Pearson's partial correlation analysis was used to estimate the correlation between cortical thickness of brain regions and DNA methylation levels of the loci. RESULTS In total, 2018 differentially methylated probes (DMPs) and 351 differentially methylated regions (DMRs) were identified. DMP-related genes were enriched in two networks involved in the central nervous system. In neuroimaging analysis, patients with MDD showed cortical thinning in the prefrontal regions and cortical thickening in several occipital regions. Cortical thickness of the left ventrolateral prefrontal cortex (VLPFC, i.e. pars triangularis) was negatively correlated with eight DMPs associated with six genes (EML6, ZFP64, CLSTN3, KCNMA1, TAOK2, and NT5E). CONCLUSION Through combining DNA methylation and neuroimaging analyses, negative correlations were identified between the cortical thickness of the left VLPFC and DNA methylation levels of eight DMPs. Our findings could improve our understanding of the pathophysiology of MDD.
Collapse
Affiliation(s)
- Hyun-Ho Yang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Kyu-Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Brain Convergence Research Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Aram Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Youbin Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo-Suk Tae
- Brain Convergence Research Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Mi-Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Brain Convergence Research Center, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Wang Z, Wu T, Hu H, Alabed AAA, Cui G, Sun L, Sun Z, Wang Y, Li P. Plasma exosomes carrying mmu-miR-146a-5p and Notch signalling pathway-mediated synaptic activity in schizophrenia. J Psychiatry Neurosci 2024; 49:E265-E281. [PMID: 39209459 PMCID: PMC11374447 DOI: 10.1503/jpn.230118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 03/13/2024] [Accepted: 05/25/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Schizophrenia is characterized by a complex interplay of genetic and environmental factors, leading to alterations in various molecular pathways that may contribute to its pathogenesis. Recent studies have shown that exosomal microRNAs could play essential roles in various brain disorders; thus, we sought to explore the potential molecular mechanisms through which microRNAs in plasma exosomes are involved in schizophrenia. METHODS We obtained sequencing data sets (SUB12404730, SUB12422862, and SUB12421357) and transcriptome sequencing data sets (GSE111708, GSE108925, and GSE18981) from mouse models of schizophrenia using the Sequence Read Archive and the Gene Expression Omnibus databases, respectively. We performed differential expression analysis on mRNA to identify differentially expressed genes. We conducted Gene Ontology (GO) functional and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses to determine differentially expressed genes. Subsequently, we determined the intersection of differentially expressed microRNAs in plasma exosomes and in prefrontal cortex tissue. We retrieved downstream target genes of mmu-miR-146a-5p from TargetScan and used Cytoscape to visualize and map the microRNA-target gene regulatory network. We conducted in vivo experiments using MK-801-induced mouse schizophrenia models and in vitro experiments using cultured mouse neurons. The role of plasma exosomal miR-146a-5p in schizophrenia was validated using a cell counting kit, detection of lactate dehydrogenase, dual-luciferase assay, quantitative reverse transcription polymerase chain reaction, and Western blot analysis. RESULTS Differential genes were mainly enriched in synaptic regulation-related functions and pathways and were associated with neuronal degeneration. We found that mmu-miR-146a-5p was highly expressed in both prefrontal cortical tissue and plasma exosomes, which may be transferred to lobe cortical vertebral neurons, leading to the synergistic dysregulation of gene network functions and, therefore, promoting schizophrenia development. We found that mmu-miR-146a-5p may inhibit the Notch signalling pathway-mediated synaptic activity of mouse pyramidal neurons in the lobe cortex by targeting NOTCH1, which in turn could promote the onset and development of schizophrenia in mice. LIMITATIONS The study's findings are based on animal models and in vitro experiments, which may not fully replicate the complexity of human schizophrenia. CONCLUSION Our findings suggest that mmu-miR-146a-5p in plasma-derived exosomes may play an important role in the pathogenesis of schizophrenia. Our results provide new insights into the underlying molecular mechanisms of the disease.
Collapse
Affiliation(s)
- Zhichao Wang
- From the Departments of Academic Research, Qiqihar Medical University, Qiqihar, PR China (Z. Wang); the School of Basic Medical Sciences, Nanchang University, Nanchang, PR China (Hu); the Community Medicine Department, Faculty of Medicine, Lincoln University College, Malaysia (Alabed); the Department of Psychology, Qiqihar Medical University, Qiqihar, PR China (Wu, Cui, L. Sun, Z. Sun)
| | - Tong Wu
- From the Departments of Academic Research, Qiqihar Medical University, Qiqihar, PR China (Z. Wang); the School of Basic Medical Sciences, Nanchang University, Nanchang, PR China (Hu); the Community Medicine Department, Faculty of Medicine, Lincoln University College, Malaysia (Alabed); the Department of Psychology, Qiqihar Medical University, Qiqihar, PR China (Wu, Cui, L. Sun, Z. Sun)
| | - Houjia Hu
- From the Departments of Academic Research, Qiqihar Medical University, Qiqihar, PR China (Z. Wang); the School of Basic Medical Sciences, Nanchang University, Nanchang, PR China (Hu); the Community Medicine Department, Faculty of Medicine, Lincoln University College, Malaysia (Alabed); the Department of Psychology, Qiqihar Medical University, Qiqihar, PR China (Wu, Cui, L. Sun, Z. Sun)
| | - Alabed Ali A Alabed
- From the Departments of Academic Research, Qiqihar Medical University, Qiqihar, PR China (Z. Wang); the School of Basic Medical Sciences, Nanchang University, Nanchang, PR China (Hu); the Community Medicine Department, Faculty of Medicine, Lincoln University College, Malaysia (Alabed); the Department of Psychology, Qiqihar Medical University, Qiqihar, PR China (Wu, Cui, L. Sun, Z. Sun)
| | - Guangcheng Cui
- From the Departments of Academic Research, Qiqihar Medical University, Qiqihar, PR China (Z. Wang); the School of Basic Medical Sciences, Nanchang University, Nanchang, PR China (Hu); the Community Medicine Department, Faculty of Medicine, Lincoln University College, Malaysia (Alabed); the Department of Psychology, Qiqihar Medical University, Qiqihar, PR China (Wu, Cui, L. Sun, Z. Sun)
| | - Lei Sun
- From the Departments of Academic Research, Qiqihar Medical University, Qiqihar, PR China (Z. Wang); the School of Basic Medical Sciences, Nanchang University, Nanchang, PR China (Hu); the Community Medicine Department, Faculty of Medicine, Lincoln University College, Malaysia (Alabed); the Department of Psychology, Qiqihar Medical University, Qiqihar, PR China (Wu, Cui, L. Sun, Z. Sun)
| | - Zhenghai Sun
- From the Departments of Academic Research, Qiqihar Medical University, Qiqihar, PR China (Z. Wang); the School of Basic Medical Sciences, Nanchang University, Nanchang, PR China (Hu); the Community Medicine Department, Faculty of Medicine, Lincoln University College, Malaysia (Alabed); the Department of Psychology, Qiqihar Medical University, Qiqihar, PR China (Wu, Cui, L. Sun, Z. Sun)
| | - Yuchen Wang
- From the Departments of Academic Research, Qiqihar Medical University, Qiqihar, PR China (Z. Wang); the School of Basic Medical Sciences, Nanchang University, Nanchang, PR China (Hu); the Community Medicine Department, Faculty of Medicine, Lincoln University College, Malaysia (Alabed); the Department of Psychology, Qiqihar Medical University, Qiqihar, PR China (Wu, Cui, L. Sun, Z. Sun)
| | - Ping Li
- From the Departments of Academic Research, Qiqihar Medical University, Qiqihar, PR China (Z. Wang); the School of Basic Medical Sciences, Nanchang University, Nanchang, PR China (Hu); the Community Medicine Department, Faculty of Medicine, Lincoln University College, Malaysia (Alabed); the Department of Psychology, Qiqihar Medical University, Qiqihar, PR China (Wu, Cui, L. Sun, Z. Sun)
| |
Collapse
|
11
|
Bou-Rouphael J, Doulazmi M, Eschstruth A, Abdou A, Durand BC. Cerebellar granular neuron progenitors exit their germinative niche via BarH-like1 activity mediated partly by inhibition of T-cell factor. Development 2024; 151:dev202234. [PMID: 38860486 DOI: 10.1242/dev.202234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Cerebellar granule neuron progenitors (GNPs) originate from the upper rhombic lip (URL), a germinative niche in which developmental defects produce human diseases. T-cell factor (TCF) responsiveness and Notch dependence are hallmarks of self-renewal in neural stem cells. TCF activity, together with transcripts encoding proneural gene repressors hairy and enhancer of split (Hes/Hey), are detected in the URL; however, their functions and regulatory modes are undeciphered. Here, we established amphibian as a pertinent model for studying vertebrate URL development. The amphibian long-lived URL is TCF active, whereas the external granular layer (EGL) is non-proliferative and expresses hes4 and hes5 genes. Using functional and transcriptomic approaches, we show that TCF activity is necessary for URL emergence and maintenance. We establish that the transcription factor Barhl1 controls GNP exit from the URL, acting partly through direct TCF inhibition. Identification of Barhl1 target genes suggests that, besides TCF, Barhl1 inhibits transcription of hes5 genes independently of Notch signaling. Observations in amniotes suggest a conserved role for Barhl in maintenance of the URL and/or EGL via co-regulation of TCF, Hes and Hey genes.
Collapse
Affiliation(s)
- Johnny Bou-Rouphael
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Mohamed Doulazmi
- Sorbonne Université, CNRS UMR8256, Institut de Biologie Paris-Seine (IBPS) - Laboratoire Adaptation Biologique et Vieillissement, 75005 Paris, France
| | - Alexis Eschstruth
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Asna Abdou
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Béatrice C Durand
- Sorbonne Université, CNRS UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, 75005 Paris, France
- Sorbonne Université, CNRS UMR8256, Institut de Biologie Paris-Seine (IBPS) - Laboratoire Adaptation Biologique et Vieillissement, 75005 Paris, France
| |
Collapse
|
12
|
Almanaa TN, Alwetaid MY, Bakheet SA, Attia SM, Ansari MA, Nadeem A, Ahmad SF. Aflatoxin B 1 exposure deteriorates immune abnormalities in a BTBR T + Itpr3 tf/J mouse model of autism by increasing inflammatory mediators' production in CD19-expressing cells. J Neuroimmunol 2024; 391:578365. [PMID: 38723577 DOI: 10.1016/j.jneuroim.2024.578365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/22/2024] [Accepted: 05/03/2024] [Indexed: 06/09/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficiencies in communication, repetitive and stereotyped behavioral patterns, and difficulties in reciprocal social engagement. The presence of immunological dysfunction in ASD has been well established. Aflatoxin B1 (AFB1) is a prevalent mycotoxin found in food and feed, causing immune toxicity and hepatotoxicity. AFB1 is significantly elevated in several regions around the globe. Existing research indicates that prolonged exposure to AFB1 results in neurological problems. The BTBR T+ Itpr3tf/J (BTBR) mice, which were used as an autism model, exhibit the primary behavioral traits that define ASD, such as repeated, stereotyped behaviors and impaired social interactions. The main objective of this work was to assess the toxic impact of AFB1 in BTBR mice. This work aimed to examine the effects of AFB1 on the expression of Notch-1, IL-6, MCP-1, iNOS, GM-CSF, and NF-κB p65 by CD19+ B cells in the spleen of the BTBR using flow cytometry. We also verified the impact of AFB1 exposure on the mRNA expression levels of Notch-1, IL-6, MCP-1, iNOS, GM-CSF, and NF-κB p65 in the brain of BTBR mice using real-time PCR. The findings of our study showed that the mice treated with AFB1 in the BTBR group exhibited a substantial increase in the presence of CD19+Notch-1+, CD19+IL-6+, CD19+MCP-1+, CD19+iNOS+, CD19+GM-CSF+, and CD19+NF-κB p65+ compared to the mice in the BTBR group that were treated with saline. Our findings also confirmed that administering AFB1 to BTBR mice leads to elevated mRNA expression levels of Notch-1, IL-6, MCP-1, iNOS, GM-CSF, and NF-κB p65 in the brain, in comparison to BTBR mice treated with saline. The data highlight that exposure to AFB1 worsens immunological abnormalities by increasing the expression of inflammatory mediators in BTBR mice.
Collapse
Affiliation(s)
- Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Y Alwetaid
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
13
|
Gyimesi M, Okolicsanyi RK, Haupt LM. Beyond amyloid and tau: rethinking Alzheimer's disease through less explored avenues. Open Biol 2024; 14:240035. [PMID: 38862019 DOI: 10.1098/rsob.240035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
Neurodegenerative diseases, particularly Alzheimer's disease (AD), pose a significant challenge in ageing populations. Our current understanding indicates that the onset of toxic amyloid and tau protein pathologies initiates disease progression. However, existing treatments targeting these hallmark symptoms offer symptomatic relief without halting disease advancement. This review offers an alternative perspective on AD, centring on impaired adult hippocampal neurogenesis (AHN) as a potential early aetiological factor. By delving into the intricate molecular events during the initial stages of AD (Braak Stages I-III), a novel hypothesis is presented, interweaving the roles of Notch signalling and heparan sulfate proteoglycans (HSPGs) in compromised AHN. While acknowledging the significance of the amyloid and tau hypotheses, it calls for further exploration beyond these paradigms, suggesting the potential of altered HS sulfation patterns in AD initiation. Future directions propose more detailed investigations into early HS aggregation, aberrant sulfation patterns and examination of their temporal relationship with tau hyperphosphorylation. In challenging the conventional 'triggers' of AD and urging their reconsideration as symptoms, this review advocates an alternative approach to understanding this disease, offering new avenues of investigation into the intricacies of AD pathogenesis.
Collapse
Affiliation(s)
- M Gyimesi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave , Kelvin Grove, Queensland 4059, Australia
| | - R K Okolicsanyi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave , Kelvin Grove, Queensland 4059, Australia
- Max Planck Queensland Centre for the Materials Sciences of Extracellular Matrices , Brisbane, QLD 4059, Australia
| | - L M Haupt
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave , Kelvin Grove, Queensland 4059, Australia
- Max Planck Queensland Centre for the Materials Sciences of Extracellular Matrices , Brisbane, QLD 4059, Australia
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), 60 Musk Ave , Kelvin Grove, Queensland 4059, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies , Brisbane, QLD 4059, Australia
| |
Collapse
|
14
|
Papageorgiou L, Papa L, Papakonstantinou E, Mataragka A, Dragoumani K, Chaniotis D, Beloukas A, Iliopoulos C, Bongcam-Rudloff E, Chrousos GP, Kossida S, Eliopoulos E, Vlachakis D. SNP and Structural Study of the Notch Superfamily Provides Insights and Novel Pharmacological Targets against the CADASIL Syndrome and Neurodegenerative Diseases. Genes (Basel) 2024; 15:529. [PMID: 38790158 PMCID: PMC11120892 DOI: 10.3390/genes15050529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
The evolutionary conserved Notch signaling pathway functions as a mediator of direct cell-cell communication between neighboring cells during development. Notch plays a crucial role in various fundamental biological processes in a wide range of tissues. Accordingly, the aberrant signaling of this pathway underlies multiple genetic pathologies such as developmental syndromes, congenital disorders, neurodegenerative diseases, and cancer. Over the last two decades, significant data have shown that the Notch signaling pathway displays a significant function in the mature brains of vertebrates and invertebrates beyond neuronal development and specification during embryonic development. Neuronal connection, synaptic plasticity, learning, and memory appear to be regulated by this pathway. Specific mutations in human Notch family proteins have been linked to several neurodegenerative diseases including Alzheimer's disease, CADASIL, and ischemic injury. Neurodegenerative diseases are incurable disorders of the central nervous system that cause the progressive degeneration and/or death of brain nerve cells, affecting both mental function and movement (ataxia). There is currently a lot of study being conducted to better understand the molecular mechanisms by which Notch plays an essential role in the mature brain. In this study, an in silico analysis of polymorphisms and mutations in human Notch family members that lead to neurodegenerative diseases was performed in order to investigate the correlations among Notch family proteins and neurodegenerative diseases. Particular emphasis was placed on the study of mutations in the Notch3 protein and the structure analysis of the mutant Notch3 protein that leads to the manifestation of the CADASIL syndrome in order to spot possible conserved mutations and interpret the effect of these mutations in the Notch3 protein structure. Conserved mutations of cysteine residues may be candidate pharmacological targets for the potential therapy of CADASIL syndrome.
Collapse
Affiliation(s)
- Louis Papageorgiou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, Agioy Spyridonos, 12243 Egaleo, Greece; (D.C.); (A.B.)
| | - Lefteria Papa
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
| | - Eleni Papakonstantinou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Antonia Mataragka
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
| | - Konstantina Dragoumani
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
| | - Dimitrios Chaniotis
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, Agioy Spyridonos, 12243 Egaleo, Greece; (D.C.); (A.B.)
| | - Apostolos Beloukas
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, Agioy Spyridonos, 12243 Egaleo, Greece; (D.C.); (A.B.)
| | - Costas Iliopoulos
- School of Informatics, Faculty of Natural & Mathematical Sciences, King’s College London, Bush House, Strand, London WC2R 2LS, UK;
| | - Erik Bongcam-Rudloff
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, 756 51 Uppsala, Sweden;
| | - George P. Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Sofia Kossida
- IMGT, The International ImMunoGenetics Information System, Laboratoire d’ImmunoGénétique Moléculaire LIGM, Institut de Génétique Humaine, (IGH), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier (UM), 34000 Montpellier, France;
| | - Elias Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
| | - Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
- School of Informatics, Faculty of Natural & Mathematical Sciences, King’s College London, Bush House, Strand, London WC2R 2LS, UK;
| |
Collapse
|
15
|
Gancheva MR, Kremer K, Breen J, Arthur A, Hamilton-Bruce A, Thomas P, Gronthos S, Koblar S. Effect of Octamer-Binding Transcription Factor 4 Overexpression on the Neural Induction of Human Dental Pulp Stem Cells. Stem Cell Rev Rep 2024; 20:797-815. [PMID: 38316679 PMCID: PMC10984899 DOI: 10.1007/s12015-024-10678-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/07/2024]
Abstract
Stem cell-based therapy is a potential alternative strategy for brain repair, with neural stem cells (NSC) presenting as the most promising candidates. Obtaining sufficient quantities of NSC for clinical applications is challenging, therefore alternative cell types, such as neural crest-derived dental pulp stem cells (DPSC), may be considered. Human DPSC possess neurogenic potential, exerting positive effects in the damaged brain through paracrine effects. However, a method for conversion of DPSC into NSC has yet to be developed. Here, overexpression of octamer-binding transcription factor 4 (OCT4) in combination with neural inductive conditions was used to reprogram human DPSC along the neural lineage. The reprogrammed DPSC demonstrated a neuronal-like phenotype, with increased expression levels of neural markers, limited capacity for sphere formation, and enhanced neuronal but not glial differentiation. Transcriptomic analysis further highlighted the expression of genes associated with neural and neuronal functions. In vivo analysis using a developmental avian model showed that implanted DPSC survived in the developing central nervous system and respond to endogenous signals, displaying neuronal phenotypes. Therefore, OCT4 enhances the neural potential of DPSC, which exhibited characteristics aligning with neuronal progenitors. This method can be used to standardise DPSC neural induction and provide an alternative source of neural cell types.
Collapse
Affiliation(s)
- Maria R Gancheva
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia.
- School of Biological Sciences, Faculty of Science, Engineering and Technology, The University of Adelaide, Adelaide, 5005, Australia.
| | - Karlea Kremer
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - James Breen
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Agnes Arthur
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Anne Hamilton-Bruce
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- Stroke Research Programme, Basil Hetzel Institute, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, 5011, Australia
| | - Paul Thomas
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide, 5000, Australia
| | - Stan Gronthos
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide, 5000, Australia
| | - Simon Koblar
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| |
Collapse
|
16
|
Wang YZ, Castillon CCM, Gebis KK, Bartom ET, d'Azzo A, Contractor A, Savas JN. Notch receptor-ligand binding facilitates extracellular vesicle-mediated neuron-to-neuron communication. Cell Rep 2024; 43:113680. [PMID: 38241148 PMCID: PMC10976296 DOI: 10.1016/j.celrep.2024.113680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/15/2023] [Accepted: 01/01/2024] [Indexed: 01/21/2024] Open
Abstract
Extracellular vesicles (EVs) facilitate intercellular communication by transferring cargo between cells in a variety of tissues. However, how EVs achieve cell-type-specific intercellular communication is still largely unknown. We found that Notch1 and Notch2 proteins are expressed on the surface of neuronal EVs that have been generated in response to neuronal excitatory synaptic activity. Notch ligands bind these EVs on the neuronal plasma membrane, trigger their internalization, activate the Notch signaling pathway, and drive the expression of Notch target genes. The generation of these neuronal EVs requires the endosomal sorting complex required for transport-associated protein Alix. Adult Alix conditional knockout mice have reduced hippocampal Notch signaling activation and glutamatergic synaptic protein expression. Thus, EVs facilitate neuron-to-neuron communication via the Notch receptor-ligand system in the brain.
Collapse
Affiliation(s)
- Yi-Zhi Wang
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Charlotte C M Castillon
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kamil K Gebis
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth T Bartom
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alessandra d'Azzo
- Department of Genetics, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anis Contractor
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jeffrey N Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
17
|
Huang L, Lai X, Liang X, Chen J, Yang Y, Xu W, Qin Q, Qin R, Huang X, Xie M, Chen L. A promise for neuronal repair: reprogramming astrocytes into neurons in vivo. Biosci Rep 2024; 44:BSR20231717. [PMID: 38175538 PMCID: PMC10830445 DOI: 10.1042/bsr20231717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/13/2023] [Accepted: 01/02/2024] [Indexed: 01/05/2024] Open
Abstract
Massive loss of neurons following brain injury or disease is the primary cause of central nervous system dysfunction. Recently, much research has been conducted on how to compensate for neuronal loss in damaged parts of the nervous system and thus restore functional connectivity among neurons. Direct somatic cell differentiation into neurons using pro-neural transcription factors, small molecules, or microRNAs, individually or in association, is the most promising form of neural cell replacement therapy available. This method provides a potential remedy for cell loss in a variety of neurodegenerative illnesses, and the development of reprogramming technology has made this method feasible. This article provides a comprehensive review of reprogramming, including the selection and methods of reprogramming starting cell populations as well as the signaling methods involved in this process. Additionally, we thoroughly examine how reprogramming astrocytes into neurons can be applied to treat stroke and other neurodegenerative diseases. Finally, we discuss the challenges of neuronal reprogramming and offer insights about the field.
Collapse
Affiliation(s)
- Lijuan Huang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xinyu Lai
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaojun Liang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Jiafeng Chen
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yue Yang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Wei Xu
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Qingchun Qin
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Rongxing Qin
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoying Huang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Minshan Xie
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Li Chen
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
18
|
Dill-McFarland KA, Altman MC, Esnault S, Jarjour NN, Busse WW, Rosenkranz MA. Molecular pathways underlying lung-brain axis signaling in asthma: Relevance for psychopathology and neuroinflammation. J Allergy Clin Immunol 2024; 153:111-121. [PMID: 37730134 PMCID: PMC10841090 DOI: 10.1016/j.jaci.2023.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Accumulating evidence indicates that asthma has systemic effects and affects brain function. Although airway inflammation is proposed to initiate afferent communications with the brain, the signaling pathways have not been established. OBJECTIVE We sought to identify the cellular and molecular pathways involved in afferent lung-brain communication during airway inflammation in asthma. METHODS In 23 adults with mild asthma, segmental bronchial provocation with allergen (SBP-Ag) was used to provoke airway inflammation and retrieve bronchoalveolar lavage fluid for targeted protein analysis and RNA sequencing to determine gene expression profiles. Neural responses to emotional cues in nodes of the salience network were assessed with functional magnetic resonance imaging at baseline and 48 hours after SBP-Ag. RESULTS Cell deconvolution and gene coexpression network analysis identified 11 cell-associated gene modules that changed in response to SBP-Ag. SBP-Ag increased bronchoalveolar lavage eosinophils and expression of an eosinophil-associated module enriched for genes related to TH17-type inflammation (eg, IL17A), as well as cell proliferation in lung and brain (eg, NOTCH1, VEGFA, and LIF). Increased expression of genes in this module, as well as several TH17-type inflammation-related proteins, was associated with an increase from baseline in salience network reactivity. CONCLUSIONS Our results identify a specific inflammatory pathway linking asthma-related airway inflammation and emotion-related neural function. Systemically, TH17-type inflammation has been implicated in both depression and neuroinflammation, with impacts on long-term brain health. Thus, our data emphasize that inflammation in the lung in asthma may have profound effects outside of the lung that may be targetable with novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Matthew C Altman
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Wash; Systems Immunology Program, Benaroya Research Institute, Seattle, Wash
| | - Stephane Esnault
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin-Madison, Madison, Wis
| | - Nizar N Jarjour
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin-Madison, Madison, Wis
| | - William W Busse
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin-Madison, Madison, Wis
| | - Melissa A Rosenkranz
- Center for Healthy Minds, University of Wisconsin-Madison, Madison, Wis; Department of Psychiatry, University of Wisconsin-Madison, Madison, Wis.
| |
Collapse
|
19
|
Wang X, Zhou R, Sun X, Li J, Wang J, Yue W, Wang L, Liu H, Shi Y, Zhang D. Preferential Regulation of Γ-Secretase-Mediated Cleavage of APP by Ganglioside GM1 Reveals a Potential Therapeutic Target for Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303411. [PMID: 37759382 PMCID: PMC10646247 DOI: 10.1002/advs.202303411] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/20/2023] [Indexed: 09/29/2023]
Abstract
A hallmark of Alzheimer's disease (AD) is the senile plaque, which contains β-amyloid peptides (Aβ). Ganglioside GM1 is the most common brain ganglioside. However, the mechanism of GM1 in modulating Aβ processing is rarely known. Aβ levels are detected by using Immunohistochemistry (IHC) and enzyme-linked immune-sorbent assay (ELISA). Cryo-electron microscopy (Cryo-EM) is used to determine the structure of γ-secretase supplemented with GM1. The levels of the cleavage of amyloid precursor protein (APP)/Cadherin/Notch1 are detected using Western blot analysis. Y maze, object translocation, and Barnes maze are performed to evaluate cognitive functions. GM1 leads to conformational change of γ-secretase structure and specifically accelerates γ-secretase cleavage of APP without affecting other substrates including Notch1, potentially through its interaction with the N-terminal fragment of presenilin 1 (PS1). Reduction of GM1 levels decreases amyloid plaque deposition and improves cognitive dysfunction. This study reveals the mechanism of GM1 in Aβ generation and provides the evidence that decreasing GM1 levels represents a potential strategy in AD treatment. These results provide insights into the detailed mechanism of the effect of GM1 on PS1, representing a step toward the characterization of its novel role in the modulation of γ-secretase activity and the pathogenesis of AD.
Collapse
Affiliation(s)
- Xiaotong Wang
- Peking University Sixth HospitalPeking University Institute of Mental HealthNHC Key Laboratory of Mental Health (Peking University)National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital)Beijing100191China
- Changping LaboratoryBeijing102206China
| | - Rui Zhou
- Beijing Frontier Research Center for Biological StructureTsinghua‐Peking Joint Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijing100084China
| | - Xiaqin Sun
- Peking University Sixth HospitalPeking University Institute of Mental HealthNHC Key Laboratory of Mental Health (Peking University)National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital)Beijing100191China
| | - Jun Li
- Peking University Sixth HospitalPeking University Institute of Mental HealthNHC Key Laboratory of Mental Health (Peking University)National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital)Beijing100191China
| | - Jinxin Wang
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain ResearchBeijing Normal UniversityBeijing100875China
| | - Weihua Yue
- Peking University Sixth HospitalPeking University Institute of Mental HealthNHC Key Laboratory of Mental Health (Peking University)National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital)Beijing100191China
- PKU‐IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Lifang Wang
- Peking University Sixth HospitalPeking University Institute of Mental HealthNHC Key Laboratory of Mental Health (Peking University)National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital)Beijing100191China
| | - Hesheng Liu
- Changping LaboratoryBeijing102206China
- Biomedical Pioneering Innovation CenterPeking UniversityBeijing100871China
| | - Yigong Shi
- Beijing Frontier Research Center for Biological StructureTsinghua‐Peking Joint Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijing100084China
- Westlake Laboratory of Life Science and BiomedicineHangzhouZhejiang310024China
- Key Laboratory of Structural Biology of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhouZhejiang310024China
- Institute of BiologyWestlake Institute for Advanced Study18 Shilongshan Road, Xihu DistrictHangzhouZhejiang310024China
| | - Dai Zhang
- Peking University Sixth HospitalPeking University Institute of Mental HealthNHC Key Laboratory of Mental Health (Peking University)National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital)Beijing100191China
- Changping LaboratoryBeijing102206China
| |
Collapse
|
20
|
Mora P, Chapouly C. Astrogliosis in multiple sclerosis and neuro-inflammation: what role for the notch pathway? Front Immunol 2023; 14:1254586. [PMID: 37936690 PMCID: PMC10627009 DOI: 10.3389/fimmu.2023.1254586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Multiple sclerosis is an autoimmune inflammatory disease of the central nervous system leading to neurodegeneration. It affects 2.3 million people worldwide, generally younger than 50. There is no known cure for the disease, and current treatment options - mainly immunotherapies to limit disease progression - are few and associated with serious side effects. In multiple sclerosis, disruption of the blood-brain barrier is an early event in the pathogenesis of lesions, predisposing to edema, excito-toxicity and inflammatory infiltration into the central nervous system. Recently, the vision of the blood brain barrier structure and integrity has changed and include contributions from all components of the neurovascular unit, among which astrocytes. During neuro-inflammation, astrocytes become reactive. They undergo morphological and molecular changes named "astrogliosis" driving the conversion from acute inflammatory injury to a chronic neurodegenerative state. Astrogliosis mechanisms are minimally explored despite their significance in regulating the autoimmune response during multiple sclerosis. Therefore, in this review, we take stock of the state of knowledge regarding astrogliosis in neuro-inflammation and highlight the central role of NOTCH signaling in the process of astrocyte reactivity. Indeed, a very detailed nomenclature published in nature neurosciences in 2021, listing all the reactive astrocyte markers fully identified in the literature, doesn't cover the NOTCH signaling. Hence, we discuss evidence supporting NOTCH1 receptor as a central regulator of astrogliosis in the pathophysiology of neuro-inflammation, notably multiple sclerosis, in human and experimental models.
Collapse
Affiliation(s)
- Pierre Mora
- Université de Bordeaux, Institut national de la santé et de la recherche médicale (INSERM), Biology of Cardiovascular Diseases, Pessac, France
| | | |
Collapse
|
21
|
Lobón-Iglesias MJ, Andrianteranagna M, Han ZY, Chauvin C, Masliah-Planchon J, Manriquez V, Tauziede-Espariat A, Turczynski S, Bouarich-Bourimi R, Frah M, Dufour C, Blauwblomme T, Cardoen L, Pierron G, Maillot L, Guillemot D, Reynaud S, Bourneix C, Pouponnot C, Surdez D, Bohec M, Baulande S, Delattre O, Piaggio E, Ayrault O, Waterfall JJ, Servant N, Beccaria K, Dangouloff-Ros V, Bourdeaut F. Imaging and multi-omics datasets converge to define different neural progenitor origins for ATRT-SHH subgroups. Nat Commun 2023; 14:6669. [PMID: 37863903 PMCID: PMC10589300 DOI: 10.1038/s41467-023-42371-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/09/2023] [Indexed: 10/22/2023] Open
Abstract
Atypical teratoid rhabdoid tumors (ATRT) are divided into MYC, TYR and SHH subgroups, suggesting diverse lineages of origin. Here, we investigate the imaging of human ATRT at diagnosis and the precise anatomic origin of brain tumors in the Rosa26-CreERT2::Smarcb1flox/flox model. This cross-species analysis points to an extra-cerebral origin for MYC tumors. Additionally, we clearly distinguish SHH ATRT emerging from the cerebellar anterior lobe (CAL) from those emerging from the basal ganglia (BG) and intra-ventricular (IV) regions. Molecular characteristics point to the midbrain-hindbrain boundary as the origin of CAL SHH ATRT, and to the ganglionic eminence as the origin of BG/IV SHH ATRT. Single-cell RNA sequencing on SHH ATRT supports these hypotheses. Trajectory analyses suggest that SMARCB1 loss induces a de-differentiation process mediated by repressors of the neuronal program such as REST, ID and the NOTCH pathway.
Collapse
Affiliation(s)
- María-Jesús Lobón-Iglesias
- INSERM U830, Laboratory of Translational Research In Pediatric Oncology, PSL Research University, SIREDO Oncology center, Institut Curie Research Center, Paris, France
| | - Mamy Andrianteranagna
- INSERM U830, Laboratory of Translational Research In Pediatric Oncology, PSL Research University, SIREDO Oncology center, Institut Curie Research Center, Paris, France
- INSERM U900, Bioinformatics, Biostatistics, Epidemiology and Computational Systems Unit, Institut Curie, Mines Paris Tech, PSL Research University, Institut Curie Research Center, Paris, France
| | - Zhi-Yan Han
- INSERM U830, Laboratory of Translational Research In Pediatric Oncology, PSL Research University, SIREDO Oncology center, Institut Curie Research Center, Paris, France
| | - Céline Chauvin
- INSERM U830, Laboratory of Translational Research In Pediatric Oncology, PSL Research University, SIREDO Oncology center, Institut Curie Research Center, Paris, France
| | - Julien Masliah-Planchon
- Somatic Genetic Unit, Department of Pathology and Diagnostic and Theranostic Medecine, Institut Curie Hospital, Paris, France
| | - Valeria Manriquez
- INSERM U932, Immunity and Cancer, PSL Research University, Institut Curie Research Center, Paris, France
| | - Arnault Tauziede-Espariat
- Department of Neuropathology, GHU Paris-Psychiatry and Neurosciences, Sainte-Anne Hospital, Paris, France
- Paris Psychiatry and Neurosciences Institute (IPNP), UMR S1266, INSERM, IMA-BRAIN, Paris, France
| | - Sandrina Turczynski
- INSERM U830, Laboratory of Translational Research In Pediatric Oncology, PSL Research University, SIREDO Oncology center, Institut Curie Research Center, Paris, France
| | - Rachida Bouarich-Bourimi
- INSERM U830, Laboratory of Translational Research In Pediatric Oncology, PSL Research University, SIREDO Oncology center, Institut Curie Research Center, Paris, France
| | - Magali Frah
- INSERM U830, Laboratory of Translational Research In Pediatric Oncology, PSL Research University, SIREDO Oncology center, Institut Curie Research Center, Paris, France
| | - Christelle Dufour
- Department of Children and Adolescents Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Thomas Blauwblomme
- Department of Pediatric Neurosurgery-AP-HP, Necker Sick Kids Hospital, Université de Paris, Paris, France
| | | | - Gaelle Pierron
- Somatic Genetic Unit, Department of Pathology and Diagnostic and Theranostic Medecine, Institut Curie Hospital, Paris, France
| | - Laetitia Maillot
- Somatic Genetic Unit, Department of Pathology and Diagnostic and Theranostic Medecine, Institut Curie Hospital, Paris, France
| | - Delphine Guillemot
- Somatic Genetic Unit, Department of Pathology and Diagnostic and Theranostic Medecine, Institut Curie Hospital, Paris, France
| | - Stéphanie Reynaud
- Somatic Genetic Unit, Department of Pathology and Diagnostic and Theranostic Medecine, Institut Curie Hospital, Paris, France
| | - Christine Bourneix
- Somatic Genetic Unit, Department of Pathology and Diagnostic and Theranostic Medecine, Institut Curie Hospital, Paris, France
| | - Célio Pouponnot
- CNRS UMR 3347, INSERM U1021, Institut Curie, PSL Research University, Université Paris-Saclay, Orsay, France
| | - Didier Surdez
- INSERM U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, SIREDO Oncology Center, Institut Curie Research Center, Paris, France
- Balgrist University Hospital, Faculty of Medicine, University of Zurich (UZH), Zurich, Switzerland
| | - Mylene Bohec
- Institut Curie, PSL University, Single Cell Initiative, ICGex Next-Generation Sequencing Platform, PSL University, 75005, Paris, France
| | - Sylvain Baulande
- Institut Curie, PSL University, Single Cell Initiative, ICGex Next-Generation Sequencing Platform, PSL University, 75005, Paris, France
| | - Olivier Delattre
- Somatic Genetic Unit, Department of Pathology and Diagnostic and Theranostic Medecine, Institut Curie Hospital, Paris, France
- INSERM U830, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, SIREDO Oncology Center, Institut Curie Research Center, Paris, France
| | - Eliane Piaggio
- INSERM U932, Immunity and Cancer, PSL Research University, Institut Curie Research Center, Paris, France
| | - Olivier Ayrault
- CNRS UMR 3347, INSERM U1021, Institut Curie, PSL Research University, Université Paris-Saclay, Orsay, France
| | - Joshua J Waterfall
- INSERM U830, Integrative Functional Genomics of Cancer Lab, PSL Research University, Institut Curie Research Center, Paris, France
- Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Nicolas Servant
- INSERM U900, Bioinformatics, Biostatistics, Epidemiology and Computational Systems Unit, Institut Curie, Mines Paris Tech, PSL Research University, Institut Curie Research Center, Paris, France
| | - Kevin Beccaria
- Department of Pediatric Neurosurgery-AP-HP, Necker Sick Kids Hospital, Université de Paris, Paris, France
| | - Volodia Dangouloff-Ros
- Pediatric Radiology Department, AP-HP, Necker Sick Kids Hospital and Paris Cite Universiy INSERM 1299 and UMR 1163, Institut Imagine, Paris, France
| | - Franck Bourdeaut
- INSERM U830, Laboratory of Translational Research In Pediatric Oncology, PSL Research University, SIREDO Oncology center, Institut Curie Research Center, Paris, France.
- Department of Pediatric Oncology, SIREDO Oncology Center, Institut Curie Hospital, Paris, and Université de Paris, Paris, France.
| |
Collapse
|
22
|
Yoshizumi A, Kuboki S, Takayashiki T, Takano S, Takayanagi R, Sonoda I, Ohtsuka M. Tspan15-ADAM10 signalling enhances cancer stem cell-like properties and induces chemoresistance via Notch1 activation in ICC. Liver Int 2023; 43:2275-2291. [PMID: 37545390 DOI: 10.1111/liv.15691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND & AIMS Notch1 activation promotes ICC progression and is associated with chemoresistance; however, therapies directly targeting Notch1 showed severe adverse effects. Notch1 activation is mediated by ADAM10, a molecular scissor that separates the target protein from its substrates in the cell membrane. Tspan15 regulates ADAM10 function, but the role of Tspan15 in ICC progression is unclear. METHODS Tspan15, ADAM10, and Notch1 expression and activation in fresh surgical specimens from 80 ICC patients and ICC cells were evaluated by immunohistochemistry, RT-PCR, western blotting, and flow cytometry. RESULTS Tspan15 expression was increased in ICC compared with adjacent liver tissue, and high Tspan15 expression was an independent factor for poor prognosis. In ICC with high Tspan15 expression, vascular invasion, lymph node metastasis, and haematogenous recurrence were increased. Tspan15 was co-expressed with ADAM10 in ICC, and associated with the expression of stemness and EMT markers. In ICC cells, Tspan15 induced ADAM10 activation by mediating the translocation of activated m-ADAM10 from the cytoplasm to the surface of the cell membrane, which further activated Notch1 by separating the intracellular domain of Notch1 from its extracellular domain, leading to enhancement of CSC-like properties and EMT. This signalling was associated with enhanced chemoresistance against gemcitabine and cisplatin. Inhibition of Tspan15 or ADAM10 is a promising therapeutic strategy in ICC, as Tspan15 or ADAM10 knockdown or treatment with ADAM10 inhibitor reduced chemoresistance and invasiveness by suppressing Notch1-mediated CSC-like properties and EMT. CONCLUSIONS Tspan15-ADAM10-Notch1 signalling is associated with aggressive tumour progression and poor prognosis in ICC.
Collapse
Affiliation(s)
- Arihito Yoshizumi
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Kuboki
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tsukasa Takayashiki
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shigetsugu Takano
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryosuke Takayanagi
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Itaru Sonoda
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masayuki Ohtsuka
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
23
|
Chen X, Bing J, Lu S, Lin S, Li H, Du S, Liu J, Xi C, Zhang X, Zeng S. Notch1 is involved in cell proliferation and neuronal differentiation in the HVC of zebra finch (Taeniopygia guttata). Behav Brain Res 2023; 452:114564. [PMID: 37459956 DOI: 10.1016/j.bbr.2023.114564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/10/2023] [Accepted: 07/01/2023] [Indexed: 07/26/2023]
Abstract
Significant sex differences are found in songbirds' song control nuclei and their controlled song behaviors. To elucidate the underlying mechanisms, we explored the role of Notch1 during the development of the high vocal centre (HVC) and song learning in zebra finch. Our study first found that Notch1 positive cells were distributed in HVC with female-biased densities at posthatching day (PHD) 15, but male-biased at PHD 45 and adult. There were about 60 putative oestrogen-responsive elements within 2.5 kb upstream of Notch1, and Notch1 mRNA in the explants that contained the developing male HVC was significantly increased after estrogen addition into the cultured medium for 48 h. After injecting Notch1-interfering lentivirus into the male or female HVC at PHD 15, cell proliferation was significantly promoted in the ventricle zone overlying the HVC at PHD 23. In addition, neuronal differentiation towards Hu+ /BrdU+ at PHD 31, mature neurons (NeuN+/BrdU+) including those projecting to RA in HVC and the sizes of HVC and RA at adult increased significantly after Notch1-interfering lentiviruses were injected into the male HVC at PHD 15. However, the above measurements decreased, following the injection of the lentiviruses expressing Notch intracellular domain (NICD). Finally, the repeat numbers of syllables 'b' or 'c' of learned songs changed after the injection of Notch1-interfering or NICD-expressing lentiviruses into the HVC at PHD15. Our study suggests that Notch1 is related to the development of HVC and song learning in the zebra finch.
Collapse
Affiliation(s)
- Xiaoning Chen
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Jie Bing
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Shan Lu
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Shiying Lin
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Hongyang Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Sanyan Du
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Jin Liu
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Chao Xi
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China
| | - Xinwen Zhang
- Hainan Instistute of Science and Technology, Haikou 571126, China; College of Life Sciences, Hainan Normal University, Haikou 571158, China.
| | - Shaoju Zeng
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
24
|
Mitsiadis TA, Pagella P, Capellini TD, Smith MM. The Notch-mediated circuitry in the evolution and generation of new cell lineages: the tooth model. Cell Mol Life Sci 2023; 80:182. [PMID: 37330998 DOI: 10.1007/s00018-023-04831-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/19/2023] [Accepted: 06/09/2023] [Indexed: 06/20/2023]
Abstract
The Notch pathway is an ancient, evolutionary conserved intercellular signaling mechanism that is involved in cell fate specification and proper embryonic development. The Jagged2 gene, which encodes a ligand for the Notch family of receptors, is expressed from the earliest stages of odontogenesis in epithelial cells that will later generate the enamel-producing ameloblasts. Homozygous Jagged2 mutant mice exhibit abnormal tooth morphology and impaired enamel deposition. Enamel composition and structure in mammals are tightly linked to the enamel organ that represents an evolutionary unit formed by distinct dental epithelial cell types. The physical cooperativity between Notch ligands and receptors suggests that Jagged2 deletion could alter the expression profile of Notch receptors, thus modifying the whole Notch signaling cascade in cells within the enamel organ. Indeed, both Notch1 and Notch2 expression are severely disturbed in the enamel organ of Jagged2 mutant teeth. It appears that the deregulation of the Notch signaling cascade reverts the evolutionary path generating dental structures more reminiscent of the enameloid of fishes rather than of mammalian enamel. Loss of interactions between Notch and Jagged proteins may initiate the suppression of complementary dental epithelial cell fates acquired during evolution. We propose that the increased number of Notch homologues in metazoa enabled incipient sister cell types to form and maintain distinctive cell fates within organs and tissues along evolution.
Collapse
Affiliation(s)
- Thimios A Mitsiadis
- Institute of Oral Biology, Centre for Dental Medicine, University of Zurich, Plattenstrasse 11, 8032, Zurich, Switzerland.
| | - Pierfrancesco Pagella
- Institute of Oral Biology, Centre for Dental Medicine, University of Zurich, Plattenstrasse 11, 8032, Zurich, Switzerland
- Wallenberg Center for Molecular Medicine (WCMM) and Department of Biomedical and Clinical Sciences, Linköpings Universitet, 581 85, Linköping, Sweden
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Moya Meredith Smith
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, King's College London, London, UK
| |
Collapse
|
25
|
Rajan A, Anhezini L, Rives-Quinto N, Chhabra JY, Neville MC, Larson ED, Goodwin SF, Harrison MM, Lee CY. Low-level repressive histone marks fine-tune gene transcription in neural stem cells. eLife 2023; 12:e86127. [PMID: 37314324 PMCID: PMC10344426 DOI: 10.7554/elife.86127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/11/2023] [Indexed: 06/15/2023] Open
Abstract
Coordinated regulation of gene activity by transcriptional and translational mechanisms poise stem cells for a timely cell-state transition during differentiation. Although important for all stemness-to-differentiation transitions, mechanistic understanding of the fine-tuning of gene transcription is lacking due to the compensatory effect of translational control. We used intermediate neural progenitor (INP) identity commitment to define the mechanisms that fine-tune stemness gene transcription in fly neural stem cells (neuroblasts). We demonstrate that the transcription factor FruitlessC (FruC) binds cis-regulatory elements of most genes uniquely transcribed in neuroblasts. Loss of fruC function alone has no effect on INP commitment but drives INP dedifferentiation when translational control is reduced. FruC negatively regulates gene expression by promoting low-level enrichment of the repressive histone mark H3K27me3 in gene cis-regulatory regions. Identical to fruC loss-of-function, reducing Polycomb Repressive Complex 2 activity increases stemness gene activity. We propose low-level H3K27me3 enrichment fine-tunes gene transcription in stem cells, a mechanism likely conserved from flies to humans.
Collapse
Affiliation(s)
- Arjun Rajan
- Life Sciences Institute, University of Michigan-Ann ArborAnn ArborUnited States
| | - Lucas Anhezini
- Life Sciences Institute, University of Michigan-Ann ArborAnn ArborUnited States
| | - Noemi Rives-Quinto
- Life Sciences Institute, University of Michigan-Ann ArborAnn ArborUnited States
| | - Jay Y Chhabra
- Life Sciences Institute, University of Michigan-Ann ArborAnn ArborUnited States
| | - Megan C Neville
- Centre for Neural Circuits and Behaviour, University of OxfordOxfordUnited Kingdom
| | - Elizabeth D Larson
- Department of Biomolecular Chemistry, University of Wisconsin-MadisonMadisonUnited States
| | - Stephen F Goodwin
- Centre for Neural Circuits and Behaviour, University of OxfordOxfordUnited Kingdom
| | - Melissa M Harrison
- Department of Biomolecular Chemistry, University of Wisconsin-MadisonMadisonUnited States
| | - Cheng-Yu Lee
- Life Sciences Institute, University of Michigan-Ann ArborAnn ArborUnited States
- Department of Cell and Developmental Biology, University of Michigan Medical SchoolAnn ArborUnited States
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan Medical SchoolAnn ArborUnited States
- Rogel Cancer Center, University of Michigan Medical SchoolAnn ArborUnited States
| |
Collapse
|
26
|
Mayor E. Neurotrophic effects of intermittent fasting, calorie restriction and exercise: a review and annotated bibliography. FRONTIERS IN AGING 2023; 4:1161814. [PMID: 37334045 PMCID: PMC10273285 DOI: 10.3389/fragi.2023.1161814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/09/2023] [Indexed: 06/20/2023]
Abstract
In the last decades, important progress has been achieved in the understanding of the neurotrophic effects of intermittent fasting (IF), calorie restriction (CR) and exercise. Improved neuroprotection, synaptic plasticity and adult neurogenesis (NSPAN) are essential examples of these neurotrophic effects. The importance in this respect of the metabolic switch from glucose to ketone bodies as cellular fuel has been highlighted. More recently, calorie restriction mimetics (CRMs; resveratrol and other polyphenols in particular) have been investigated thoroughly in relation to NSPAN. In the narrative review sections of this manuscript, recent findings on these essential functions are synthesized and the most important molecules involved are presented. The most researched signaling pathways (PI3K, Akt, mTOR, AMPK, GSK3β, ULK, MAPK, PGC-1α, NF-κB, sirtuins, Notch, Sonic hedgehog and Wnt) and processes (e.g., anti-inflammation, autophagy, apoptosis) that support or thwart neuroprotection, synaptic plasticity and neurogenesis are then briefly presented. This provides an accessible entry point to the literature. In the annotated bibliography section of this contribution, brief summaries are provided of about 30 literature reviews relating to the neurotrophic effects of interest in relation to IF, CR, CRMs and exercise. Most of the selected reviews address these essential functions from the perspective of healthier aging (sometimes discussing epigenetic factors) and the reduction of the risk for neurodegenerative diseases (Alzheimer's disease, Huntington's disease, Parkinson's disease) and depression or the improvement of cognitive function.
Collapse
|
27
|
Linesch PW, Akhtar AA, Breunig JJ. Tetracycline-Inducible and Reversible Stable Gene Expression in Human iPSC-Derived Neural Progenitors and in the Postnatal Mouse Brain. Curr Protoc 2023; 3:e792. [PMID: 37283517 PMCID: PMC10264152 DOI: 10.1002/cpz1.792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Our group has developed several approaches for stable, non-viral integration of inducible transgenic elements into the genome of mammalian cells. Specifically, a piggyBac tetracycline-inducible genetic element of interest (pB-tet-GOI) plasmid system allows for stable piggyBac transposition-mediated integration into cells, identification of cells that have been transfected using a fluorescent nuclear reporter, and robust transgene activation or suppression upon the addition of doxycycline (dox) to the cell culture or the diet of the animal. Furthermore, the addition of luciferase downstream of the target gene allows for quantitative assessment of gene activity in a non-invasive manner. More recently, we have developed a transgenic system as an alternative to piggyBac called mosaic analysis by dual recombinase-mediated cassette exchange (MADR), as well as additional in vitro transfection techniques and in vivo dox chow applications. The protocols herein provide instructions for the use of this system in cell lines and in the neonatal mouse brain. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Cloning of respective genetic element of interest (GOI) into response plasmid Basic Protocol 2: In vitro nucleofection of iPSC-derived human/mouse neural progenitor cells and subsequent derivation of stable inducible cell lines Alternate Protocol: In vitro electroporation of iPSC-derived human/mouse neural progenitor cells Support Protocol: Recovery stage after in vitro transfection Basic Protocol 3: Adding doxycycline to cells to induce/reverse GOI Basic Protocol 4: Assessing gene expression in vitro by non-invasive bioluminescence imaging of luciferase activity.
Collapse
Affiliation(s)
- Paul W. Linesch
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Medicine, UCLA, Los Angeles, California
| |
Collapse
|
28
|
Shao L, Paik N, Sanborn M, Bandara T, Vijaykumar A, Sottoriva K, Rehman J, Nombela-Arrieta C, Pajcini K. Hematopoietic Jagged1 is a fetal liver niche factor required for functional maturation and engraftment of fetal hematopoietic stem cells. Proc Natl Acad Sci U S A 2023; 120:e2210058120. [PMID: 37155858 PMCID: PMC10193977 DOI: 10.1073/pnas.2210058120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 04/04/2023] [Indexed: 05/10/2023] Open
Abstract
Notch signaling is essential for the emergence of definitive hematopoietic stem cells (HSCs) in the embryo and their development in the fetal liver niche. However, how Notch signaling is activated and which fetal liver cell type provides the ligand for receptor activation in HSCs is unknown. Here we provide evidence that endothelial Jagged1 (Jag1) has a critical early role in fetal liver vascular development but is not required for hematopoietic function during fetal HSC expansion. We demonstrate that Jag1 is expressed in many hematopoietic cells in the fetal liver, including HSCs, and that its expression is lost in adult bone marrow HSCs. Deletion of hematopoietic Jag1 does not affect fetal liver development; however, Jag1-deficient fetal liver HSCs exhibit a significant transplantation defect. Bulk and single-cell transcriptomic analysis of HSCs during peak expansion in the fetal liver indicates that loss of hematopoietic Jag1 leads to the downregulation of critical hematopoietic factors such as GATA2, Mllt3, and HoxA7, but does not perturb Notch receptor expression. Ex vivo activation of Notch signaling in Jag1-deficient fetal HSCs partially rescues the functional defect in a transplant setting. These findings indicate a new fetal-specific niche that is based on juxtracrine hematopoietic Notch signaling and reveal Jag1 as a fetal-specific niche factor essential for HSC function.
Collapse
Affiliation(s)
- Lijian Shao
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| | - Na Yoon Paik
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| | - Mark A. Sanborn
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL60612
| | - Thilinie Bandara
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| | - Anjali Vijaykumar
- Department of Medical Oncology and Hematology, University Hospital Zurich, 8091Zurich, Switzerland
| | - Kilian Sottoriva
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL60612
| | - Cesar Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital Zurich, 8091Zurich, Switzerland
| | - Kostandin V. Pajcini
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| |
Collapse
|
29
|
Devanand M, V N S, Madhu K. Signaling mechanisms involved in the regulation of remyelination in multiple sclerosis: a mini review. J Mol Med (Berl) 2023:10.1007/s00109-023-02312-9. [PMID: 37084092 DOI: 10.1007/s00109-023-02312-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 02/22/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Multiple sclerosis is an autoimmune neurodegenerative disease of the CNS that causes progressive disabilities, owing to CNS axon degeneration as a late result of demyelination. In the search for the prevention of axonal loss, mitigating inflammatory attacks in the CNS and myelin restoration are two possible approaches. As a result, therapies that target diverse signaling pathways involved in neuroprotection and remyelination have the potential to overcome the challenges in the development of multiple sclerosis treatments. LINGO1 (Leucine rich repeat and Immunoglobulin domain containing, Nogo receptor- interaction protein), AKT/PIP3/mTOR, Notch, Wnt, RXR (Retinoid X receptor gamma), and Nrf2 (nuclear factor erythroid 2-related factor 2) signaling pathways are highlighted in this section. This article reviews the present knowledge regarding numerous signaling pathways and their functions in regulating remyelination in multiple sclerosis pathogenesis. These pathways are potential biomarkers and therapeutic targets in MS.
Collapse
Affiliation(s)
- Midhuna Devanand
- Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Saiprabha V N
- Department of Pharmaceutical Chemistry and Analysis, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| | - Krishnadas Madhu
- Department of Pharmacology, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
30
|
Regulatory role of melatonin in Notch1 signaling pathway in cerebral cortex of Aβ 1-42-induced Alzheimer's disease rat model. Mol Biol Rep 2023; 50:2463-2469. [PMID: 36602704 DOI: 10.1007/s11033-022-08213-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Soluble Amyloid-beta (Aβ) oligomers are thought to play a key role in the pathogenesis of Alzheimer's disease (AD), which is the most common age-associated neurodegenerative diseases with obvious neuropathological changes and functional decline in both cortical and subcortical regions. Melatonin is ubiquitously distributed and multifunctioning indoleamine. Accumulating studies support that melatonin is potential therapeutic molecule for AD through modulating a broad variety of signaling pathways. In recent years, Notch1 signaling pathway is been known involved in dynamic changes in the cellular architecture and function of adult brain, as well as associated with the pathophysiology of AD and other neurodegenerative disorders. METHODS AND RESULTS In this study, we performed real-time polymerase chain reaction, immunohistochemistry and western blotting analyses using the cerebral cortical tissues of Aβ1-42 oligomers-induced AD rats with or without melatonin treatment. Our results showed that soluble Aβ1-42 oligomers decreased the expression of the main components of Notch1 signaling pathway, Notch1, NICD and Hes1 in the cerebral cortex, and melatonin could restore the level of Notch1, NICD and Hes1. CONCLUSION This observation suggests that targeting of Notch1 signaling might be a promising therapeutic approach for AD and other age-associated neurodegenerative diseases, and melatonin might serve as a potential therapeutic agent for AD and other age-associated neurodegenerative diseases.
Collapse
|
31
|
Comparisons between Plant and Animal Stem Cells Regarding Regeneration Potential and Application. Int J Mol Sci 2023; 24:ijms24054392. [PMID: 36901821 PMCID: PMC10002278 DOI: 10.3390/ijms24054392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Regeneration refers to the process by which organisms repair and replace lost tissues and organs. Regeneration is widespread in plants and animals; however, the regeneration capabilities of different species vary greatly. Stem cells form the basis for animal and plant regeneration. The essential developmental processes of animals and plants involve totipotent stem cells (fertilized eggs), which develop into pluripotent stem cells and unipotent stem cells. Stem cells and their metabolites are widely used in agriculture, animal husbandry, environmental protection, and regenerative medicine. In this review, we discuss the similarities and differences in animal and plant tissue regeneration, as well as the signaling pathways and key genes involved in the regulation of regeneration, to provide ideas for practical applications in agriculture and human organ regeneration and to expand the application of regeneration technology in the future.
Collapse
|
32
|
Soto M, Fernández M, Bravo P, Lahoz S, Garrido A, Sánchez-Rodríguez A, Rivera-Sánchez M, Sierra M, Melón P, Roig-García A, Naito A, Casey B, Camps J, Tolosa E, Martí MJ, Infante J, Ezquerra M, Fernández-Santiago R. Differential serum microRNAs in premotor LRRK2 G2019S carriers from Parkinson's disease. NPJ Parkinsons Dis 2023; 9:15. [PMID: 36732514 PMCID: PMC9894906 DOI: 10.1038/s41531-023-00451-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 01/09/2023] [Indexed: 02/04/2023] Open
Abstract
The LRRK2 G2019S pathogenic mutation causes LRRK2-associated Parkinson's disease (L2PD) with incomplete penetrance. LRRK2 non-manifesting carriers (L2NMC) are at PD high risk but predicting pheno-conversion is challenging given the lack of progression biomarkers. To investigate novel biomarkers for PD premotor stages, we performed a longitudinal microRNA (miRNA) assessment of serum samples from G2019S L2NMC followed-up over 8 years. Our cohort consisted of G2019S L2NMC stratified by dopamine transporter single-photon emission computed tomography (DaT-SPECT) into DaT-negative (n = 20) and DaT-positive L2NMC (n = 20), pheno-converted G2019S L2PD patients (n = 20), idiopathic PD (iPD) (n = 19), and controls (n = 40). We also screened a second cohort of L2PD patients (n = 19) and controls (n = 20) (Total n = 158). Compared to healthy controls, we identified eight deregulated miRNAs in DaT-negative L2NMC, six in DaT-positive L2NMC, and one in L2PD. Between groups, the highest miRNA differences, 24 candidate miRNAs, occurred between DaT-positive L2NMC and L2PD. Longitudinally, we found 11 common miRNAs with sustained variation in DaT-negative and DaT-positive L2NMCs compared to their baselines. Our study identifies novel miRNA alterations in premotor stages of PD co-occurring with progressive DaT-SPECT decline before motor manifestation, whose deregulation seems to attenuate after the diagnosis of L2PD. Moreover, we identified four miRNAs with relatively high discriminative ability (AUC = 0.82) between non-pheno-converted DaT-positive G2019S carriers and pheno-converted L2PD patients (miR-4505, miR-8069, miR-6125, and miR-451a), which hold potential as early progression biomarkers for PD.
Collapse
Affiliation(s)
- Marta Soto
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain
| | - Manel Fernández
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain
- Parkinson's Disease and Movement Disorders Group of the Institut de Neurociències (Universitat de Barcelona), ES-08036, Barcelona, Catalonia, Spain
| | - Paloma Bravo
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain
| | - Sara Lahoz
- Gastrointestinal and Pancreatic Oncology Team, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic de Barcelona, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Alicia Garrido
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain
| | - Antonio Sánchez-Rodríguez
- Movement Disorders Unit, Department of Neurology, Hospital Universitario Marqués de Valdecilla, Universidad de Cantabria, ES-39008, Santander, Cantabria, Spain
| | - María Rivera-Sánchez
- Movement Disorders Unit, Department of Neurology, Hospital Universitario Marqués de Valdecilla, Universidad de Cantabria, ES-39008, Santander, Cantabria, Spain
| | - María Sierra
- Movement Disorders Unit, Department of Neurology, Hospital Universitario Marqués de Valdecilla, Universidad de Cantabria, ES-39008, Santander, Cantabria, Spain
| | - Paula Melón
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain
| | - Ana Roig-García
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain
| | - Anna Naito
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station, P.O. Box 4777, New York, NY, 10120, USA
| | - Bradford Casey
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station, P.O. Box 4777, New York, NY, 10120, USA
| | - Jordi Camps
- Gastrointestinal and Pancreatic Oncology Team, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic de Barcelona, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Eduardo Tolosa
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain
| | - María-José Martí
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain
| | - Jon Infante
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain
- Movement Disorders Unit, Department of Neurology, Hospital Universitario Marqués de Valdecilla, Universidad de Cantabria, ES-39008, Santander, Cantabria, Spain
| | - Mario Ezquerra
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain.
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain.
| | - Rubén Fernández-Santiago
- Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain.
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, ES-08036, Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), ES-08036, Barcelona, Catalonia, Spain.
- Histology Unit, Department of Biomedicine, Faculty of Medicine, Universitat de Barcelona, ES-08036, Barcelona, Catalonia, Spain.
| |
Collapse
|
33
|
Abdi IY, Bartl M, Dakna M, Abdesselem H, Majbour N, Trenkwalder C, El-Agnaf O, Mollenhauer B. Cross-sectional proteomic expression in Parkinson's disease-related proteins in drug-naïve patients vs healthy controls with longitudinal clinical follow-up. Neurobiol Dis 2023; 177:105997. [PMID: 36634823 DOI: 10.1016/j.nbd.2023.105997] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/11/2023] Open
Abstract
There is an urgent need to find reliable and accessible blood-based biomarkers for early diagnosis of Parkinson's disease (PD) correlating with clinical symptoms and displaying predictive potential to improve future clinical trials. This led us to a conduct large-scale proteomics approach using an advanced high-throughput proteomics technology to create a proteomic profile for PD. Over 1300 proteins were measured in serum samples from a de novo Parkinson's (DeNoPa) cohort made up of 85 deep clinically phenotyped drug-naïve de novo PD patients and 93 matched healthy controls (HC) with longitudinal clinical follow-up available of up to 8 years. The analysis identified 73 differentially expressed proteins (DEPs) of which 14 proteins were confirmed as stable potential diagnostic markers using machine learning tools. Among the DEPs identified, eight proteins-ALCAM, contactin 1, CD36, DUS3, NEGR1, Notch1, TrkB, and BTK- significantly correlated with longitudinal clinical scores including motor and non-motor symptom scores, cognitive function and depression scales, indicating potential predictive values for progression in PD among various phenotypes. Known functions of these proteins and their possible relation to the pathophysiology or symptomatology of PD were discussed and presented with a particular emphasis on the potential biological mechanisms involved, such as cell adhesion, axonal guidance and neuroinflammation, and T-cell activation. In conclusion, with the use of advance multiplex proteomic technology, a blood-based protein signature profile was identified from serum samples of a well-characterized PD cohort capable of potentially differentiating PD from HC and predicting clinical disease progression of related motor and non-motor PD symptoms. We thereby highlight the need to validate and further investigate these markers in future prospective cohorts and assess their possible PD-related mechanisms.
Collapse
Affiliation(s)
- Ilham Yahya Abdi
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, Doha, Qatar; Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| | - Michael Bartl
- Department of Neurology, University Medical Center Goettingen, Robert-Koch, Goettingen, Germany.
| | - Mohammed Dakna
- Department of Neurology, University Medical Center Goettingen, Robert-Koch, Goettingen, Germany.
| | - Houari Abdesselem
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| | - Nour Majbour
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Klinikstrasse, Kassel, Germany; Department of Neurosurgery, University Medical Center Goettingen, Robert-Koch, Goettingen, Germany.
| | - Omar El-Agnaf
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, Doha, Qatar; Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Robert-Koch, Goettingen, Germany; Paracelsus-Elena-Klinik, Klinikstrasse, Kassel, Germany.
| |
Collapse
|
34
|
Zhu C, Zhang L, Heidari M, Sun S, Chang S, Xie Q, Ai Y, Dong K, Zhang H. Small RNA deep sequencing revealed microRNAs' involvement in modulating cellular senescence and immortalization state. Poult Sci 2023; 102:102474. [PMID: 36689784 PMCID: PMC9876980 DOI: 10.1016/j.psj.2022.102474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/01/2022] [Accepted: 12/31/2022] [Indexed: 01/05/2023] Open
Abstract
Unlike rodent cells, spontaneous immortalization of avian cells and human cells is a very rare event. According to patent publications and current literature, there are no more than 4 spontaneously immortalized chicken embryo fibroblast (CEF) cell lines established up to date. One of those cell lines is ADOL (Avian Disease and Oncology Laboratory) ZS-1 cell line, which was established by continuous passaging of the CEFs derived from the specific pathogen free (SPF) 0.TVB*S1 (commonly known as rapid feathering susceptible or RFS) genetic line of chickens. The RFS genetic line of chickens was developed and has been maintained on the SPF chicken farm of USDA-ARS facility, ADOL, in East Lansing, Michigan, which is known as one of a few lines of chickens that are free of any known avian endogenous virus genes. To explore potential roles that epigenetic factors may play in modulating cellular senescence processes and spontaneous immortalization state, total RNAs extracted from samples of the RFS primary CEFs, RFS CEFs reached the 21st passage, and the ZS-1 cells were subjected to small RNA sequencing. Collectively, a total of 531 miRNAs was identified in the 3 types of samples. In contrast to the primary CEF samples, 50 miRNAs were identified with significantly differential expression only in the 21st passage samples; a different subset of 63 differentially expressed miRNAs was identified only in the ZS-1 samples; the majority of differentially expressed miRNAs identified in both the 21st passage CEF and the ZS-1 samples were more or less directionally consistent. Gene Ontology analysis results suggested that the epigenetic factor, miRNAs, plays a role in modulating the cellular senescence and spontaneous immortalization processes through various bioprocesses and key pathways including ErbB and MAPK signaling pathways. These findings provided the experimental and bioinformatic evidence for a better understanding on the epigenetic factor of miRNAs in association with cellular senescence and spontaneous immortalization process in avian cells.
Collapse
Affiliation(s)
- Chen Zhu
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA,Michigan State University, East Lansing, MI 48824, USA
| | - Lei Zhang
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA,Institute of Special Wild Economic Animal and Plant Science, Chinese Academy of Agricultural Sciences, Changchun, Jilin 130112, China
| | - Mohammad Heidari
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA
| | - Shuhong Sun
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Shuang Chang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Qingmei Xie
- College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yongxing Ai
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Kunzhe Dong
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912, USA
| | - Huanmin Zhang
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA.
| |
Collapse
|
35
|
Conith AJ, Hope SA, Albertson RC. Covariation of brain and skull shapes as a model to understand the role of crosstalk in development and evolution. Evol Dev 2023; 25:85-102. [PMID: 36377237 PMCID: PMC9839637 DOI: 10.1111/ede.12421] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/24/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022]
Abstract
Covariation among discrete phenotypes can arise due to selection for shared functions, and/or shared genetic and developmental underpinnings. The consequences of such phenotypic integration are far-reaching and can act to either facilitate or limit morphological variation. The vertebrate brain is known to act as an "organizer" of craniofacial development, secreting morphogens that can affect the shape of the growing neurocranium, consistent with roles for pleiotropy in brain-neurocranium covariation. Here, we test this hypothesis in cichlid fishes by first examining the degree of shape integration between the brain and the neurocranium using three-dimensional geometric morphometrics in an F5 hybrid population, and then genetically mapping trait covariation using quantitative trait loci (QTL) analysis. We observe shape associations between the brain and the neurocranium, a pattern that holds even when we assess associations between the brain and constituent parts of the neurocranium: the rostrum and braincase. We also recover robust genetic signals for both hard- and soft-tissue traits and identify a genomic region where QTL for the brain and braincase overlap, implicating a role for pleiotropy in patterning trait covariation. Fine mapping of the overlapping genomic region identifies a candidate gene, notch1a, which is known to be involved in patterning skeletal and neural tissues during development. Taken together, these data offer a genetic hypothesis for brain-neurocranium covariation, as well as a potential mechanism by which behavioral shifts may simultaneously drive rapid change in neuroanatomy and craniofacial morphology.
Collapse
Affiliation(s)
- Andrew J. Conith
- Biology DepartmentUniversity of Massachusetts AmherstAmherstMassachusettsUSA
| | - Sylvie A. Hope
- Biology DepartmentUniversity of Massachusetts AmherstAmherstMassachusettsUSA
| | - R. Craig Albertson
- Biology DepartmentUniversity of Massachusetts AmherstAmherstMassachusettsUSA
| |
Collapse
|
36
|
Barth K, Vasić V, McDonald B, Heinig N, Wagner MC, Schumann U, Röhlecke C, Bicker F, Schumann L, Radyushkin K, Baumgart J, Tenzer S, Zipp F, Meinhardt M, Alitalo K, Tegeder I, Schmidt MHH. EGFL7 loss correlates with increased VEGF-D expression, upregulating hippocampal adult neurogenesis and improving spatial learning and memory. Cell Mol Life Sci 2023; 80:54. [PMID: 36715759 PMCID: PMC9886625 DOI: 10.1007/s00018-023-04685-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/31/2023]
Abstract
Neural stem cells reside in the subgranular zone, a specialized neurogenic niche of the hippocampus. Throughout adulthood, these cells give rise to neurons in the dentate gyrus, playing an important role in learning and memory. Given that these core cognitive processes are disrupted in numerous disease states, understanding the underlying mechanisms of neural stem cell proliferation in the subgranular zone is of direct practical interest. Here, we report that mature neurons, neural stem cells and neural precursor cells each secrete the neurovascular protein epidermal growth factor-like protein 7 (EGFL7) to shape this hippocampal niche. We further demonstrate that EGFL7 knock-out in a Nestin-CreERT2-based mouse model produces a pronounced upregulation of neurogenesis within the subgranular zone. RNA sequencing identified that the increased expression of the cytokine VEGF-D correlates significantly with the ablation of EGFL7. We substantiate this finding with intraventricular infusion of VEGF-D upregulating neurogenesis in vivo and further show that VEGF-D knock-out produces a downregulation of neurogenesis. Finally, behavioral studies in EGFL7 knock-out mice demonstrate greater maintenance of spatial memory and improved memory consolidation in the hippocampus by modulation of pattern separation. Taken together, our findings demonstrate that both EGFL7 and VEGF-D affect neurogenesis in the adult hippocampus, with the ablation of EGFL7 upregulating neurogenesis, increasing spatial learning and memory, and correlating with increased VEGF-D expression.
Collapse
Affiliation(s)
- Kathrin Barth
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr. 74, 01307 Dresden, Germany
| | - Verica Vasić
- Institute of Medical Informatics and Biometry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany ,Institute of Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Brennan McDonald
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr. 74, 01307 Dresden, Germany
| | - Nora Heinig
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr. 74, 01307 Dresden, Germany
| | - Marc-Christoph Wagner
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr. 74, 01307 Dresden, Germany ,Institute of Medical Informatics and Biometry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany
| | - Ulrike Schumann
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr. 74, 01307 Dresden, Germany
| | - Cora Röhlecke
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr. 74, 01307 Dresden, Germany
| | - Frank Bicker
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany ,Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lana Schumann
- Institute of Clinical Pharmacology, Goethe-University Hospital Frankfurt Am Main, Frankfurt, Germany
| | - Konstantin Radyushkin
- Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany ,Mouse Behavior Outcome Unit, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jan Baumgart
- Translational Animal Research Center (TARC), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Tenzer
- Institute of Immunology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany ,Focus Program Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany ,Focus Program Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany ,Department of Neurology, Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Meinhardt
- Institute of Pathology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kari Alitalo
- Translational Cancer Medicine Program and iCAN Digital Precision Cancer Medicine Flagship, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Hospital Frankfurt Am Main, Frankfurt, Germany
| | - Mirko H. H. Schmidt
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr. 74, 01307 Dresden, Germany
| |
Collapse
|
37
|
Zamfirescu AM, Yatsenko AS, Shcherbata HR. Notch signaling sculpts the stem cell niche. Front Cell Dev Biol 2022; 10:1027222. [PMID: 36605720 PMCID: PMC9810114 DOI: 10.3389/fcell.2022.1027222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Adult stem cells depend on their niches for regulatory signaling that controls their maintenance, division, and their progeny differentiation. While communication between various types of stem cells and their niches is becoming clearer, the process of stem cell niche establishment is still not very well understood. Model genetic organisms provide simplified systems to address various complex questions, for example, how is a stem cell niche formed? What signaling cascades induce the stem cell niche formation? Are the mechanisms of stem cell niche formation conserved? Notch signaling is an evolutionarily conserved pathway first identified in fruit flies, crucial in fate acquisition and spatiotemporal patterning. While the core logic behind its activity is fairly simple and requires direct cell-cell interaction, it reaches an astonishing complexity and versatility by combining its different modes of action. Subtleties such as equivalency between communicating cells, their physical distance, receptor and ligand processing, and endocytosis can have an effect on the way the events unfold, and this review explores some important general mechanisms of action, later on focusing on its involvement in stem cell niche formation. First, looking at invertebrates, we will examine how Notch signaling induces the formation of germline stem cell niche in male and female Drosophila. In the developing testis, a group of somatic gonadal precursor cells receive Delta signals from the gut, activating Notch signaling and sealing their fate as niche cells even before larval hatching. Meanwhile, the ovarian germline stem cell niche is built later during late larval stages and requires a two-step process that involves terminal filament formation and cap cell specification. Intriguingly, double security mechanisms of Notch signaling activation coordinated by the soma or the germline control both steps to ensure the robustness of niche assembly. Second, in the vast domains of mammalian cellular signaling, there is an emerging picture of Notch being an active player in a variety of tissues in health and disease. Notch involvement has been shown in stem cell niche establishment in multiple organs, including the brain, muscle, and intestine, where the stem cell niches are essential for the maintenance of adult stem cells. But adult stem cells are not the only cells looking for a home. Cancer stem cells use Notch signaling at specific stages to gain an advantage over endogenous tissue and overpower it, at the same time acquiring migratory and invasive abilities to claim new tissues (e.g., bone) as their territory. Moreover, in vitro models such as organoids reveal similar Notch employment when it comes to the developing stem cell niches. Therefore, a better understanding of the processes regulating stem cell niche assembly is key for the fields of stem cell biology and regenerative medicines.
Collapse
Affiliation(s)
| | | | - Halyna R. Shcherbata
- Mount Desert Island Biological Laboratory, Bar Harbor, ME, United States,*Correspondence: Halyna R. Shcherbata,
| |
Collapse
|
38
|
Kosa P, Barbour C, Varosanec M, Wichman A, Sandford M, Greenwood M, Bielekova B. Molecular models of multiple sclerosis severity identify heterogeneity of pathogenic mechanisms. Nat Commun 2022; 13:7670. [PMID: 36509784 PMCID: PMC9744737 DOI: 10.1038/s41467-022-35357-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
While autopsy studies identify many abnormalities in the central nervous system (CNS) of subjects dying with neurological diseases, without their quantification in living subjects across the lifespan, pathogenic processes cannot be differentiated from epiphenomena. Using machine learning (ML), we searched for likely pathogenic mechanisms of multiple sclerosis (MS). We aggregated cerebrospinal fluid (CSF) biomarkers from 1305 proteins, measured blindly in the training dataset of untreated MS patients (N = 129), into models that predict past and future speed of disability accumulation across all MS phenotypes. Healthy volunteers (N = 24) data differentiated natural aging and sex effects from MS-related mechanisms. Resulting models, validated (Rho 0.40-0.51, p < 0.0001) in an independent longitudinal cohort (N = 98), uncovered intra-individual molecular heterogeneity. While candidate pathogenic processes must be validated in successful clinical trials, measuring them in living people will enable screening drugs for desired pharmacodynamic effects. This will facilitate drug development making, it hopefully more efficient and successful.
Collapse
Affiliation(s)
- Peter Kosa
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Christopher Barbour
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Mihael Varosanec
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Alison Wichman
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Mary Sandford
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Mark Greenwood
- grid.41891.350000 0001 2156 6108Department of Mathematical Sciences, Montana State University, Bozeman, MT USA
| | - Bibiana Bielekova
- grid.94365.3d0000 0001 2297 5165Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
39
|
Kwak M, Southard KM, Kim WR, Lin A, Kim NH, Gopalappa R, Lee HJ, An M, Choi SH, Jung Y, Noh K, Farlow J, Georgakopoulos A, Robakis NK, Kang MK, Kutys ML, Seo D, Kim HH, Kim YH, Cheon J, Gartner ZJ, Jun YW. Adherens junctions organize size-selective proteolytic hotspots critical for Notch signalling. Nat Cell Biol 2022; 24:1739-1753. [PMID: 36456828 PMCID: PMC10665132 DOI: 10.1038/s41556-022-01031-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/19/2022] [Indexed: 12/02/2022]
Abstract
Adherens junctions (AJs) create spatially, chemically and mechanically discrete microdomains at cellular interfaces. Here, using a mechanogenetic platform that generates artificial AJs with controlled protein localization, clustering and mechanical loading, we find that AJs also organize proteolytic hotspots for γ-secretase with a spatially regulated substrate selectivity that is critical in the processing of Notch and other transmembrane proteins. Membrane microdomains outside of AJs exclusively organize Notch ligand-receptor engagement (LRE microdomains) to initiate receptor activation. Conversely, membrane microdomains within AJs exclusively serve to coordinate regulated intramembrane proteolysis (RIP microdomains). They do so by concentrating γ-secretase and primed receptors while excluding full-length Notch. AJs induce these functionally distinct microdomains by means of lipid-dependent γ-secretase recruitment and size-dependent protein segregation. By excluding full-length Notch from RIP microdomains, AJs prevent inappropriate enzyme-substrate interactions and suppress spurious Notch activation. Ligand-induced ectodomain shedding eliminates size-dependent segregation, releasing Notch to translocate into AJs for processing by γ-secretase. This mechanism directs radial differentiation of ventricular zone-neural progenitor cells in vivo and more broadly regulates the proteolysis of other large cell-surface receptors such as amyloid precursor protein. These findings suggest an unprecedented role of AJs in creating size-selective spatial switches that choreograph γ-secretase processing of multiple transmembrane proteins regulating development, homeostasis and disease.
Collapse
Affiliation(s)
- Minsuk Kwak
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
| | - Kaden M Southard
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Woon Ryoung Kim
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Annie Lin
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Nam Hyeong Kim
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
- Department of Nano Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Imnewrun Inc., Suwon, Republic of Korea
| | - Ramu Gopalappa
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Lee
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Minji An
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Seo Hyun Choi
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Yunmin Jung
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Kunwoo Noh
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Justin Farlow
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Anastasios Georgakopoulos
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikolaos K Robakis
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Min K Kang
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew L Kutys
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Daeha Seo
- Department of Physics and Chemistry, DGIST, Daegu, Republic of Korea
| | - Hyongbum Henry Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Ho Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
- Department of Nano Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Imnewrun Inc., Suwon, Republic of Korea
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul, Republic of Korea
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Young-Wook Jun
- Department of Otolaryngology, University of California, San Francisco, CA, USA.
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
40
|
Hashimoto M, Takeichi K, Murata K, Kozakai A, Yagi A, Ishikawa K, Suzuki-Nakagawa C, Kasuya Y, Fukamizu A, Nakagawa T. Regulation of neural stem cell proliferation and survival by protein arginine methyltransferase 1. Front Neurosci 2022; 16:948517. [PMID: 36440275 PMCID: PMC9685794 DOI: 10.3389/fnins.2022.948517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/17/2022] [Indexed: 12/22/2024] Open
Abstract
Protein arginine methyltransferase 1 (PRMT1), a major type I arginine methyltransferase in mammals, methylates histone and non-histone proteins to regulate various cellular functions, such as transcription, DNA damage response, and signal transduction. PRMT1 is highly expressed in neural stem cells (NSCs) and embryonic brains, suggesting that PRMT1 is essential for early brain development. Although our previous reports have shown that PRMT1 positively regulates oligodendrocyte development, it has not been studied whether PRMT1 regulates NSC proliferation and its survival during development. To examine the role of PRMT1 in NSC activity, we cultured NSCs prepared from embryonic mouse forebrains deficient in PRMT1 specific for NSCs and performed neurosphere assays. We found that the primary neurospheres of PRMT1-deficient NSCs were small and the number of spheres was decreased, compared to those of control NSCs. Primary neurospheres deficient in PRMT1 expressed an increased level of cleaved caspase-3, suggesting that PRMT1 deficiency-induced apoptosis. Furthermore, p53 protein was significantly accumulated in PRMT1-deficient NSCs. In parallel, p53-responsive pro-apoptotic genes including Pmaip1 and Perp were upregulated in PRMT1-deficient NSCs. p53-target p21 mRNA and its protein levels were shown to be upregulated in PRMT1-deficient NSCs. Moreover, the 5-bromo-2'-deoxyuridine (BrdU) incorporation assay showed that the loss of PRMT1 led to cell cycle defects in the embryonic NSCs. In contrast to the above in vitro observations, NSCs normally proliferated and survived in the fetal brains of NSC-specific PRMT1-deficient mice. We also found that Lama1, which encodes the laminin subunit α1, was significantly upregulated in the embryonic brains of PRMT1-deficient mice. These data implicate that extracellular factors provided by neighboring cells in the microenvironment gave a trophic support to NSCs in the PRMT1-deficient brain and recovered NSC activity to maintain brain homeostasis. Our study implies that PRMT1 plays a cell-autonomous role in the survival and proliferation of embryonic NSCs.
Collapse
Affiliation(s)
- Misuzu Hashimoto
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Kaho Takeichi
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Kazuya Murata
- Laboratory Animal Resource Center in Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Aoi Kozakai
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Atsushi Yagi
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Kohei Ishikawa
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Chiharu Suzuki-Nakagawa
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Yoshitoshi Kasuya
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
- World Premier International Research Center Initiative, International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Tsutomu Nakagawa
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
41
|
Luo H, Li M, Wang F, Yang Y, Wang Q, Zhao Y, Du F, Chen Y, Shen J, Zhao Q, Zeng J, Wang S, Chen M, Li X, Li W, Sun Y, Gu L, Wen Q, Xiao Z, Wu X. The role of intestinal stem cell within gut homeostasis: Focusing on its interplay with gut microbiota and the regulating pathways. Int J Biol Sci 2022; 18:5185-5206. [PMID: 35982910 PMCID: PMC9379405 DOI: 10.7150/ijbs.72600] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/29/2022] [Indexed: 12/05/2022] Open
Abstract
Intestinal stem cells (ISCs) play an important role in maintaining intestinal homeostasis via promoting a healthy gut barrier. Within the stem cell niche, gut microbiota linking the crosstalk of dietary influence and host response has been identified as a key regulator of ISCs. Emerging insights from recent research reveal that ISC and gut microbiota interplay regulates epithelial self-renewal. This article reviews the recent knowledge on the key role of ISC in their local environment (stem cell niche) associating with gut microbiota and their metabolites as well as the signaling pathways. The current progress of intestinal organoid culture is further summarized. Subsequently, the key challenges and future directions are discussed.
Collapse
Affiliation(s)
- Haoming Luo
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Yifei Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Qin Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou 646000, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou 646000, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou 646000, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou 646000, Sichuan, China
| | - Qianyun Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Jiuping Zeng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Qinglian Wen
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Zhangang Xiao
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
42
|
Wang J, Chen S, Pan C, Li G, Tang Z. Application of Small Molecules in the Central Nervous System Direct Neuronal Reprogramming. Front Bioeng Biotechnol 2022; 10:799152. [PMID: 35875485 PMCID: PMC9301571 DOI: 10.3389/fbioe.2022.799152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
The lack of regenerative capacity of neurons leads to poor prognoses for some neurological disorders. The use of small molecules to directly reprogram somatic cells into neurons provides a new therapeutic strategy for neurological diseases. In this review, the mechanisms of action of different small molecules, the approaches to screening small molecule cocktails, and the methods employed to detect their reprogramming efficiency are discussed, and the studies, focusing on neuronal reprogramming using small molecules in neurological disease models, are collected. Future research efforts are needed to investigate the in vivo mechanisms of small molecule-mediated neuronal reprogramming under pathophysiological states, optimize screening cocktails and dosing regimens, and identify safe and effective delivery routes to promote neural regeneration in different neurological diseases.
Collapse
Affiliation(s)
| | | | | | - Gaigai Li
- *Correspondence: Gaigai Li, ; Zhouping Tang,
| | | |
Collapse
|
43
|
Nagase Y, Hiramatsu K, Funauchi M, Shiomi M, Masuda T, Kakuda M, Nakagawa S, Miyoshi A, Matsuzaki S, Kobayashi E, Kimura T, Serada S, Ueda Y, Naka T, Kimura T. Anti-lipolysis-stimulated lipoprotein receptor monoclonal antibody as a novel therapeutic agent for endometrial cancer. BMC Cancer 2022; 22:679. [PMID: 35729527 PMCID: PMC9210735 DOI: 10.1186/s12885-022-09789-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/16/2022] [Indexed: 12/29/2022] Open
Abstract
Background Endometrial cancer (EC) is a common gynecologic malignancy and patients with advanced and recurrent EC have a poor prognosis. Although chemotherapy is administered for those patients, the efficacy of current chemotherapy is limited. Therefore, it is necessary to develop novel therapeutic agents for EC. In this study, we focused on lipolysis-stimulated lipoprotein receptor (LSR), a membrane protein highly expressed in EC cells, and developed a chimeric chicken–mouse anti-LSR monoclonal antibody (mAb). This study investigated the antitumor effect of an anti-LSR mAb and the function of LSR in EC. Methods We examined the expression of LSR in 228 patients with EC using immunohistochemistry and divided them into two groups: high-LSR (n = 153) and low-LSR groups (n = 75). We developed a novel anti-LSR mAb and assessed its antitumor activity in an EC cell xenograft mouse model. Pathway enrichment analysis was performed using protein expression data of EC samples. LSR-knockdown EC cell lines (HEC1 and HEC116) were generated by transfected with small interfering RNA and used for assays in vitro. Results High expression of LSR was associated with poor overall survival (hazard ratio: 3.53, 95% confidence interval: 1.35–9.24, p = 0.01), advanced stage disease (p = 0.045), deep myometrial invasion (p = 0.045), and distant metastasis (p < 0.01). In EC with deep myometrial invasion, matrix metalloproteinase (MMP) 2 was highly expressed along with LSR. Anti-LSR mAb significantly inhibited the tumor growth in EC cell xenograft mouse model (tumor volume, 407.1 mm3versus 726.3 mm3, p = 0.019). Pathway enrichment analysis identified the mitogen-activated protein kinase (MAPK) pathway as a signaling pathway associated with LSR expression. Anti-LSR mAb suppressed the activity of MAPK in vivo. In vitro assays using EC cell lines demonstrated that LSR regulated cell proliferation, invasion, and migration through MAPK signaling, particularly MEK/ERK signaling and membrane-type 1 MMP (MT1-MMP) and MMP2. Moreover, ERK1/2-knockdown suppressed cell proliferation, invasion, migration, and the expression of MT1-MMP and MMP2. Conclusions Our results suggest that LSR contributes to tumor growth, invasion, metastasis, and poor prognosis of EC through MAPK signaling. Anti-LSR mAb is a potential therapeutic agent for EC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09789-6.
Collapse
Affiliation(s)
- Yoshikazu Nagase
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kosuke Hiramatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Masashi Funauchi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Division of Clinical Immunology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan.,Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Iwate, Japan.,Department of Clinical Immunology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Mayu Shiomi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tatsuo Masuda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mamoru Kakuda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Nakagawa
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ai Miyoshi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shinya Matsuzaki
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Gynecology, Osaka International Cancer Institute, Osaka, Japan
| | - Eiji Kobayashi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshihiro Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Serada
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Iwate, Japan.,Department of Clinical Immunology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tetsuji Naka
- Division of Clinical Immunology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan.,Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Iwate, Japan.,Department of Clinical Immunology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
44
|
Willems S, Marschner JA, Kilu W, Faudone G, Busch R, Duensing‐Kropp S, Heering J, Merk D. Nurr1 Modulation Mediates Neuroprotective Effects of Statins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104640. [PMID: 35488520 PMCID: PMC9218776 DOI: 10.1002/advs.202104640] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/15/2022] [Indexed: 05/09/2023]
Abstract
The ligand-sensing transcription factor Nurr1 emerges as a promising therapeutic target for neurodegenerative pathologies but Nurr1 ligands for functional studies and therapeutic validation are lacking. Here pronounced Nurr1 modulation by statins for which clinically relevant neuroprotective effects are demonstrated, is reported. Several statins directly affect Nurr1 activity in cellular and cell-free settings with low micromolar to sub-micromolar potencies. Simvastatin as example exhibits anti-inflammatory effects in astrocytes, which are abrogated by Nurr1 knockdown. Differential gene expression analysis in native and Nurr1-silenced cells reveals strong proinflammatory effects of Nurr1 knockdown while simvastatin treatment induces several neuroprotective mechanisms via Nurr1 involving changes in inflammatory, metabolic and cell cycle gene expression. Further in vitro evaluation confirms reduced inflammatory response, improved glucose metabolism, and cell cycle inhibition of simvastatin-treated neuronal cells. These findings suggest Nurr1 involvement in the well-documented but mechanistically elusive neuroprotection by statins.
Collapse
Affiliation(s)
- Sabine Willems
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 9Frankfurt60438Germany
| | - Julian A. Marschner
- Department of PharmacyLudwig‐Maximilians‐Universität MünchenButenandtstr. 5‐13Munich81377Germany
| | - Whitney Kilu
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 9Frankfurt60438Germany
| | - Giuseppe Faudone
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 9Frankfurt60438Germany
| | - Romy Busch
- Department of PharmacyLudwig‐Maximilians‐Universität MünchenButenandtstr. 5‐13Munich81377Germany
| | - Silke Duensing‐Kropp
- Department of PharmacyLudwig‐Maximilians‐Universität MünchenButenandtstr. 5‐13Munich81377Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPTheodor‐Stern‐Kai 7Frankfurt60596Germany
| | - Daniel Merk
- Institute of Pharmaceutical ChemistryGoethe University FrankfurtMax‐von‐Laue‐Str. 9Frankfurt60438Germany
- Department of PharmacyLudwig‐Maximilians‐Universität MünchenButenandtstr. 5‐13Munich81377Germany
| |
Collapse
|
45
|
Im DS, Joselin A, Svoboda D, Takano T, Rousseaux MWC, Callaghan S, Slack RS, Hisanaga SI, Davis RJ, Park DS, Qu D. Cdk5-mediated JIP1 phosphorylation regulates axonal outgrowth through Notch1 inhibition. BMC Biol 2022; 20:115. [PMID: 35581583 PMCID: PMC9115922 DOI: 10.1186/s12915-022-01312-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 04/26/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Activated Cdk5 regulates a number of processes during nervous system formation, including neuronal differentiation, growth cone stabilization, and axonal growth. Cdk5 phosphorylates its downstream substrates located in axonal growth cones, where the highly expressed c-Jun N-terminal kinase (JNK)-interacting protein1 (JIP1) has been implicated as another important regulator of axonal growth. In addition, stringent control of the level of intracellular domain of Notch1 (Notch1-IC) plays a regulatory role in axonal outgrowth during neuronal differentiation. However, whether Cdk5-JIP1-Notch1 cooperate to regulate axonal outgrowth, and the mechanism of such joint contribution to this pathway, is presently unknown, and here we explore their potential interaction. RESULTS Our interactome screen identified JIP1 as an interactor of p35, a Cdk5 activator, and we sought to explore the relationship between Cdk5 and JIP1 on the regulation of axonal outgrowth. We demonstrate that JIP1 phosphorylated by Cdk5 at Thr205 enhances axonal outgrowth and a phosphomimic JIP1 rescues the axonal outgrowth defects in JIP1-/- and p35-/- neurons. Axonal outgrowth defects caused by the specific increase of Notch1 in JIP1-/- neurons are rescued by Numb-mediated inhibition of Notch1. Finally, we demonstrate that Cdk5 phosphorylation of JIP1 further amplifies the phosphorylation status of yet another Cdk5 substrate E3-ubiquitin ligase Itch, resulting in increased Notch1 ubiquitination. CONCLUSIONS Our findings identify a potentially critical signaling axis involving Cdk5-JIP1-Itch-Notch1, which plays an important role in the regulation of CNS development. Future investigation into the way this pathway integrates with additional pathways regulating axonal growth will further our knowledge of normal central nervous system development and pathological conditions.
Collapse
Affiliation(s)
- Doo Soon Im
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Alvin Joselin
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Devon Svoboda
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Tesuya Takano
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Maxime W C Rousseaux
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Steve Callaghan
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Shin-Ichi Hisanaga
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01650, USA
| | - David S Park
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| | - Dianbo Qu
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
46
|
Xia Y, Zhang Y, Xu M, Zou X, Gao J, Ji MH, Chen G. Presenilin enhancer 2 is crucial for the transition of apical progenitors into neurons but into not basal progenitors in the developing hippocampus. Development 2022; 149:275418. [PMID: 35575074 DOI: 10.1242/dev.200272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 05/04/2022] [Indexed: 12/23/2022]
Abstract
Recent evidence has shown that presenilin enhancer 2 (Pen2; Psenen) plays an essential role in corticogenesis by regulating the switch of apical progenitors (APs) to basal progenitors (BPs). The hippocampus is a brain structure required for advanced functions, including spatial navigation, learning and memory. However, it remains unknown whether Pen2 is important for hippocampal morphogenesis. To address this question, we generated Pen2 conditional knockout (cKO) mice, in which Pen2 is inactivated in neural progenitor cells (NPCs) in the hippocampal primordium. We showed that Pen2 cKO mice exhibited hippocampal malformation and decreased population of NPCs in the neuroepithelium of the hippocampus. We found that deletion of Pen2 neither affected the proliferative capability of APs nor the switch of APs to BPs in the hippocampus, and that it caused enhanced transition of APs to neurons. We demonstrated that expression of the Notch1 intracellular domain (N1ICD) significantly increased the population of NPCs in the Pen2 cKO hippocampus. Collectively, this study uncovers a crucial role for Pen2 in the maintenance of NPCs during hippocampal development.
Collapse
Affiliation(s)
- Yingqian Xia
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu, China, 210061
| | - Yizhi Zhang
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu, China, 210061
| | - Min Xu
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, Jiangsu, China, 211166
| | - Xiaochuan Zou
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu, China, 210061
| | - Jun Gao
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, Jiangsu, China, 211166
| | - Mu-Huo Ji
- Department of Anesthesiology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China, 210003
| | - Guiquan Chen
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu, China, 210061.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China, 226001
| |
Collapse
|
47
|
Hu Y, Fang X, Wang J, Ren TT, Zhao YY, Dai JF, Qin XY, Lan R. Astragalin attenuates AlCl3/D-galactose-induced aging-like disorders by inhibiting oxidative stress and neuroinflammation. Neurotoxicology 2022; 91:60-68. [DOI: 10.1016/j.neuro.2022.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/17/2022] [Accepted: 05/04/2022] [Indexed: 12/21/2022]
|
48
|
α-Arrestins and Their Functions: From Yeast to Human Health. Int J Mol Sci 2022; 23:ijms23094988. [PMID: 35563378 PMCID: PMC9105457 DOI: 10.3390/ijms23094988] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/10/2022] Open
Abstract
α-Arrestins, also called arrestin-related trafficking adaptors (ARTs), constitute a large family of proteins conserved from yeast to humans. Despite their evolutionary precedence over their extensively studied relatives of the β-arrestin family, α-arrestins have been discovered relatively recently, and thus their properties are mostly unexplored. The predominant function of α-arrestins is the selective identification of membrane proteins for ubiquitination and degradation, which is an important element in maintaining membrane protein homeostasis as well as global cellular metabolisms. Among members of the arrestin clan, only α-arrestins possess PY motifs that allow canonical binding to WW domains of Rsp5/NEDD4 ubiquitin ligases and the subsequent ubiquitination of membrane proteins leading to their vacuolar/lysosomal degradation. The molecular mechanisms of the selective substrate’s targeting, function, and regulation of α-arrestins in response to different stimuli remain incompletely understood. Several functions of α-arrestins in animal models have been recently characterized, including redox homeostasis regulation, innate immune response regulation, and tumor suppression. However, the molecular mechanisms of α-arrestin regulation and substrate interactions are mainly based on observations from the yeast Saccharomyces cerevisiae model. Nonetheless, α-arrestins have been implicated in health disorders such as diabetes, cardiovascular diseases, neurodegenerative disorders, and tumor progression, placing them in the group of potential therapeutic targets.
Collapse
|
49
|
Tamai S, Ichinose T, Tsutsui T, Tanaka S, Garaeva F, Sabit H, Nakada M. Tumor Microenvironment in Glioma Invasion. Brain Sci 2022; 12:brainsci12040505. [PMID: 35448036 PMCID: PMC9031400 DOI: 10.3390/brainsci12040505] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/05/2023] Open
Abstract
A major malignant trait of gliomas is their remarkable infiltration capacity. When glioma develops, the tumor cells have already reached the distant part. Therefore, complete removal of the glioma is impossible. Recently, research on the involvement of the tumor microenvironment in glioma invasion has advanced. Local hypoxia triggers cell migration as an environmental factor. The transcription factor hypoxia-inducible factor (HIF) -1α, produced in tumor cells under hypoxia, promotes the transcription of various invasion related molecules. The extracellular matrix surrounding tumors is degraded by proteases secreted by tumor cells and simultaneously replaced by an extracellular matrix that promotes infiltration. Astrocytes and microglia become tumor-associated astrocytes and glioma-associated macrophages/microglia, respectively, in relation to tumor cells. These cells also promote glioma invasion. Interactions between glioma cells actively promote infiltration of each other. Surgery, chemotherapy, and radiation therapy transform the microenvironment, allowing glioma cells to invade. These findings indicate that the tumor microenvironment may be a target for glioma invasion. On the other hand, because the living body actively promotes tumor infiltration in response to the tumor, it is necessary to reconsider whether the invasion itself is friend or foe to the brain.
Collapse
|
50
|
Ho MT, Lu J, Vazquez-Pianzola P, Suter B. α-Phenylalanyl tRNA synthetase competes with Notch signaling through its N-terminal domain. PLoS Genet 2022; 18:e1010185. [PMID: 35486661 PMCID: PMC9094542 DOI: 10.1371/journal.pgen.1010185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 05/11/2022] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
Abstract
The alpha subunit of the cytoplasmic Phenylalanyl tRNA synthetase (α-PheRS, FARSA in humans) displays cell growth and proliferation activities and its elevated levels can induce cell fate changes and tumor-like phenotypes that are neither dependent on the canonical function of charging tRNAPhe with phenylalanine nor on stimulating general translation. In intestinal stem cells of Drosophila midguts, α-PheRS levels are naturally slightly elevated and human FARSA mRNA levels are elevated in multiple cancers. In the Drosophila midgut model, elevated α-PheRS levels caused the accumulation of many additional proliferating cells resembling intestinal stem cells (ISCs) and enteroblasts (EBs). This phenotype partially resembles the tumor-like phenotype described as Notch RNAi phenotype for the same cells. Genetic interactions between α-PheRS and Notch suggest that their activities neutralize each other and that elevated α-PheRS levels attenuate Notch signaling when Notch induces differentiation into enterocytes, type II neuroblast stem cell proliferation, or transcription of a Notch reporter. These non-canonical functions all map to the N-terminal part of α-PheRS which accumulates naturally in the intestine. This truncated version of α-PheRS (α-S) also localizes to nuclei and displays weak sequence similarity to the Notch intracellular domain (NICD), suggesting that α-S might compete with the NICD for binding to a common target. Supporting this hypothesis, the tryptophan (W) residue reported to be key for the interaction between the NICD and the Su(H) BTD domain is not only conserved in α-PheRS and α-S, but also essential for attenuating Notch signaling. Aminoacyl tRNA synthetases charge tRNAs with their cognate amino acid to ensure proper decoding of the genetic code during translation. Independent of its aminoacylation function, the alpha subunit of Drosophila cytoplasmic Phenylalanyl tRNA synthetase (α-PheRS, FARSA in humans) has an additional activity that promotes growth and proliferation. Here we describe that elevated α-PheRS levels also induce cell fate changes and tumorous phenotypes in Drosophila midguts. Excessive proliferating cells with stem and progenitor cell characteristics accumulate and the composition of the terminally differentiated cells changes, too. This phenotype together with observed genetic interactions between α-PheRS and Notch levels show that α-PheRS counteracts Notch signaling in many different tissues and developmental stages. This novel activity of α-PheRS maps to its N-terminal part, which is naturally produced. The fragment contains a DNA binding domain, translocates into nuclei, and displays essential similarities to a Notch domain that binds to the downstream transcription factor. This suggests that it might be competing with Notch for binding to a common target. Not only because Notch plays important roles in many tumors, but also because FARSA mRNA levels are considerably upregulated in many tumors, this novel activity deserves more attention for cancer research.
Collapse
Affiliation(s)
- Manh Tin Ho
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Jiongming Lu
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | | | - Beat Suter
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|